US20080183395A1 - Gene expression profiling for identification, monitoring and treatment of multiple sclerosis - Google Patents

Gene expression profiling for identification, monitoring and treatment of multiple sclerosis Download PDF

Info

Publication number
US20080183395A1
US20080183395A1 US11/827,892 US82789207A US2008183395A1 US 20080183395 A1 US20080183395 A1 US 20080183395A1 US 82789207 A US82789207 A US 82789207A US 2008183395 A1 US2008183395 A1 US 2008183395A1
Authority
US
United States
Prior art keywords
subject
multiple sclerosis
data set
gene expression
gene
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/827,892
Inventor
Michael Bevilacqua
Victor Tryon
Danute Bankaitis-Davis
Lisa Siconolfi
David B. Trollinger
Karl Wassmann
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Life Technologies Corp
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/821,850 external-priority patent/US6692916B2/en
Priority claimed from US10/291,225 external-priority patent/US6960439B2/en
Priority claimed from US10/742,458 external-priority patent/US20050060101A1/en
Priority claimed from US11/155,930 external-priority patent/US20060115826A1/en
Application filed by Individual filed Critical Individual
Priority to US11/827,892 priority Critical patent/US20080183395A1/en
Publication of US20080183395A1 publication Critical patent/US20080183395A1/en
Assigned to SOURCE PRECISION MEDICINE, INC. D/B/A SOURCE MDX reassignment SOURCE PRECISION MEDICINE, INC. D/B/A SOURCE MDX ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SICONOLFI, LISA, TROLLINGER, DAVID B., BANKAITIS-DAVIS, DANUTE, WASSMANN, KARL, BEVILACQUA, MICHAEL, TRYON, VICTOR
Assigned to Life Technologies Corporation reassignment Life Technologies Corporation ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SOURCE PRECISION MEDICINE, INC. D/B/A SOURCE MDX
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6888Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for detection or identification of organisms
    • C12Q1/689Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for detection or identification of organisms for bacteria
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/525Tumor necrosis factor [TNF]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/285Demyelinating diseases; Multipel sclerosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B25/00ICT specially adapted for hybridisation; ICT specially adapted for gene or protein expression

Definitions

  • the present invention relates generally to the identification of biological markers associated with the identification of multiple sclerosis. More specifically, the invention relates to the use of gene expression data in the identification, monitoring and treatment of subjects receiving anti-TNF therapy.
  • flare-ups also called relapses, attacks, or exacerbations. These are episodes of acute worsening of neurologic function. They are followed by partial or complete recovery periods (remissions) free of disease progression.
  • the invention is based in part upon the identification of gene expression profiles associated with multiple sclerosis (MS). Theses genes are referred to herein as MS-associated genes. More specifically, the invention is based upon the surprising discovery that detection of as few as two MS-associated genes is capable of identifying individuals with or without MS with at least 75% accuracy.
  • MS multiple sclerosis
  • the invention provides a method for determining a profile data set for characterizing a subject with multiple sclerosis or an inflammatory condition related to multiple sclerosis based on a sample from the subject, the sample providing a source of RNAs, by using amplification for measuring the amount of RNA in a panel of constituents including at least 2 constituents from any of Tables 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 and arriving at a measure of each constituent.
  • the profile data set contains the measure of each constituent of the panel.
  • Also provided by the invention is a method of characterizing multiple sclerosis or inflammatory condition related to multiple sclerosis in a subject, based on a sample from the subject, the sample providing a source of RNAs, by assessing a profile data set of a plurality of members, each member being a quantitative measure of the amount of a distinct RNA constituent in a panel of constituents selected so that measurement of the constituents enables characterization of the presumptive signs of a multiple sclerosis.
  • the invention provides a method of characterizing multiple sclerosis or an inflammatory condition related to multiple sclerosis in a subject, based on a sample from the subject, the sample providing a source of RNAs, by determining a quantitative measure of the amount of at least one constituent from any of Tables 1-10.
  • the invention provides a method for predicting an adverse effect from anti-TNF therapy in a subject, based on a sample from the subject, the sample providing a source of RNAs, said method comprising: a) assessing a profile data set of a plurality of members, each member being a quantitative measure of the amount of a distinct RNA constituent in a panel of constituents selected so that measurement of the constituents enables characterization of the presumptive signs of multiple sclerosis or an inflammatory condition related to multiple sclerosis, wherein such measure for each constituent is obtained under measurement conditions that are substantially repeatable to produce a patient data set; and b) comparing the patient data set to a baseline profile data set, wherein the baseline profile data set is related to said multiple sclerosis or inflammatory condition related to multiple sclerosis; wherein a similarity between the patient data set and the baseline profile data set indicates a risk of an adverse effect from anti-TNF therapy in the subject.
  • the present invention provides a method for predicting an increased risk to an adverse effect from anti-TNF therapy in a subject, based on a sample from the subject, the sample providing a source of RNAs, said method comprising: a) determining a quantitative measure of the amount of at least one constituent of Table 4 or 10 as a distinct RNA constituent, wherein such measure is obtained under measurement conditions that are substantially repeatable to produce a patient data set; and b) comparing the patient data set to a baseline profile data set, wherein the baseline profile data set is related to said multiple sclerosis or inflammatory condition related to multiple sclerosis; wherein a similarity between the patient data set and the baseline profile data set indicates a risk of an adverse effect from anti-TNF therapy in the subject.
  • the method of predicting an adverse effect of anti-TNF therapy is performed on a subject suffering from an inflammatory condition, including but not limited to rheumatoid arthritis, psoriasis, ankylosing spondylitis, psoriatic arthritis and Crohn's diseases.
  • the method is performed prior to, during, or after administering an anti-TNF therapeutic or anti-TNF therapeutic regimen to the subject.
  • said method is performed prior to administering an anti-TNF therapeutic to the subject.
  • increased risk it is meant that treatment with anti-TNF therapy is contraindicated.
  • the panel of constituents are selected so as to distinguish from a normal and a MS-diagnosed subject.
  • the MS-diagnosed subject is washed out from therapy for three or more months.
  • the panel of constituents are selected so as to distinguish from a normal and a MS-diagnosed subject with at least 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or greater accuracy.
  • accuracy is meant that the method has the ability to distinguish between subjects having multiple sclerosis or an inflammatory condition associated with multiple sclerosis and those that do not. Accuracy is determined for example by comparing the results of the Gene Expression Profiling to standard accepted clinical methods of diagnosing MS, e.g. MRI, sign and symptoms such as blurred vision, fatigue, loss or balance.
  • the panel of constituents is selected as to permit characterizing severity of MS in relation to normal over time so as to track movement toward normal as a result of successful therapy and away from normal in response to symptomatic flare.
  • the panel contains 10, 8, 5, 4, 3 or fewer constituents.
  • the panel of constituents includes ITGAM, HLADRA, CASP9, ITGAL or STAT3.
  • the panel includes ITGAM and i) CD4 and MMP9, ii) ITGA4 and MMP9, iii) ITGA4, MMP9 and CALCA, iv) ITGA4, MMP9 and NFKB1B, v) ITGA4, MMP9, CALCA and CXCR3, or vi) ITGA4, MMP9, NFKB1B and CXCR3.
  • the panel includes two or more constituents from any of Tables 1-10. In one preferred embodiment, the panel includes two or more constituents from Table 4 or 10.
  • the panel includes three constituents in any combination shown on Table 7.
  • the panel includes any 2, 3, 4, or 5 genes in the combination shown in Tables 6, 7, 8 and 9 respectively.
  • the panel contains i) HLADRA and one or more or the following: ITGAL, CASP9, NFKB1B, STAT2, NFKB1, ITGAM, ITGAL, CD4, IL1B, HSPA1A, ICAM1, IFI16, or TGFBR2; ii) CASP9 and one or more of the following VEGFB, CD14 or JUN; iii) ITGAL and one or more of the following: P13, ITGAM or TGFBR2; and iv) STAT3 and CD14.
  • assessing may further include comparing the profile data set to a baseline profile data set for the panel.
  • the baseline profile data set is related to the multiple sclerosis or an inflammatory condition related to multiple sclerosis to be characterized.
  • the baseline profile data set is derived from one or more other samples from the same subject, taken when the subject is in a biological condition different from that in which the subject was at the time the first sample was taken, with respect to at least one of age, nutritional history, medical condition, clinical indicator, medication, physical activity, body mass, and environmental exposure, and the baseline profile data set may be derived from one or more other samples from one or more different subjects.
  • the one or more different subjects may have in common with the subject at least one of age group, gender, ethnicity, geographic location, nutritional history, medical condition, clinical indicator, medication, physical activity, body mass, and environmental exposure.
  • a clinical indicator may be used to assess multiple sclerosis or am inflammatory condition related to multiple sclerosis of the one or more different subjects, and may also include interpreting the calibrated profile data set in the context of at least one other clinical indicator, wherein the at least one other clinical indicator such as blood chemistry, urinalysis, X-ray or other radiological or metabolic imaging technique, other chemical assays, and physical findings.
  • the baseline profile data set may be derived from one or more other samples from the same subject taken under circumstances different from those of the first sample, and the circumstances may be selected from the group consisting of (i) the time at which the first sample is taken, (ii) the site from which the first sample is taken, (iii) the biological condition of the subject when the first sample is taken.
  • Presumptive signs of multiple sclerosis includes for example, altered sensory, motor, visual or proprioceptive system with at least one of numbness or weakness in one or more limbs, often occurring on one side of the body at a time or the lower half of the body, partial or complete loss of vision, frequently in one eye at a time and often with pain during eye movement, double vision or blurring of vision, tingling or pain in numb areas of the body, electric-shock sensations that occur with certain head movements, tremor, lack of coordination or unsteady gait, fatigue, dizziness, muscle stiffness or spasticity, slurred speech, paralysis, problems with bladder, bowel or sexual function, and mental changes such as forgetfulness or difficulties with concentration, relative to medical standards.
  • the subject is at risk of developing multiple sclerosis, for example the subject has a family history of multiple sclerosis or another autoimmune disorder such as for example rheumatoid arthritis, Crohn's disease, or lupus.
  • subject is a candidate for anti-TNF therapy.
  • multiple sclerosis or an inflammatory condition related to multiple sclerosis is meant that the condition is an autoimmune condition, an environmental condition, a viral infection, a bacterial infection, a eukaryotic parasitic infection, or a fungal infection.
  • the sample is any sample derived from a subject which contains RNA.
  • the sample is blood, a blood fraction, body fluid, and a population of cells or tissue from the subject.
  • one or more other samples can be taken over an interval of time that is at least one month between the first sample and the one or more other samples, or taken over an interval of time that is at least twelve months between the first sample and the one or more samples, or they may be taken pre-therapy intervention or post-therapy intervention.
  • the first sample may be derived from blood and the baseline profile data set may be derived from tissue or body fluid of the subject other than blood.
  • the first sample is derived from tissue or body fluid of the subject and the baseline profile data set is derived from blood.
  • the measurement conditions are substantially repeatable, particularly within a degree of repeatability of better than five percent or more particularly within a degree of repeatability of better than three percent, and/or wherein efficiencies of amplification for all constituents are substantially similar, more particularly wherein the efficiency of amplification is within two percent, and still more particularly wherein the efficiency of amplification for all constituents is less than one percent.
  • the invention includes storing the profile data set in a digital storage medium.
  • storing the profile data set includes storing it as a record in a database.
  • FIG. 1A shows the results of assaying 24 genes from the Source Inflammation Gene Panel (shown in Table 1 of U.S. Pat. No. 6,692,916, which patent is hereby incorporated by reference; such Panel is hereafter referred to as the Inflammation Gene Expression Panel, and is incorporated into the 96 gene expression panel shown in Table 10, referred to as the Precision ProfileTM for Inflammatory Response) on eight separate days during the course of optic neuritis in a single male subject.
  • the Source Inflammation Gene Panel shown in Table 1 of U.S. Pat. No. 6,692,916, which patent is hereby incorporated by reference; such Panel is hereafter referred to as the Inflammation Gene Expression Panel, and is incorporated into the 96 gene expression panel shown in Table 10, referred to as the Precision ProfileTM for Inflammatory Response
  • FIG. 1 B illustrates use of an inflammation index in relation to the data of FIG. 1A , in accordance with an embodiment of the present invention.
  • FIG. 2 is a graphical illustration of the same inflammation index calculated at 9 different, significant clinical milestones.
  • FIG. 3 shows the effects of single dose treatment with 800 mg of ibuprofen in a single donor as characterized by the index.
  • FIG. 4 shows the calculated acute inflammation index displayed graphically for five different conditions.
  • FIG. 5 shows a Viral Response Index for monitoring the progress of an upper respiratory infection (URI).
  • URI upper respiratory infection
  • FIGS. 6 and 7 compare two different populations using Gene Expression Profiles (with respect to the 48 loci of the Inflammation Gene Expression Panel (which is incorporated in the Precision ProfileTM for Inflammatory Response shown in Table 10).
  • FIG. 8 compares a normal population with a rheumatoid arthritis population derived from a longitudinal study.
  • FIG. 9 compares two normal populations, one longitudinal and the other cross sectional.
  • FIG. 10 shows the shows gene expression values for various individuals of a normal population.
  • FIG. 11 shows the expression levels for each of four genes of the Inflammation Gene Expression Panel (which is incorporated in the Precision ProfileTM for Inflammatory Response shown in Table 10), of a single subject, assayed monthly over a period of eight months.
  • FIG. 12 shows the expression levels for each of 48 genes of the Inflammation Gene Expression Panel, (which is incorporated in the Precision ProfileTM for Inflammatory Response shown in Table 10) of distinct single subjects (selected in each case on the basis of feeling well and not taking drugs), assayed weekly over a period of four weeks.
  • FIG. 13 shows the expression levels for each of 48 genes (of the Inflammation Gene Expression Panel (which is incorporated in the Precision ProfileTM for Inflammatory Response shown in Table 10), of distinct single subjects (selected in each case on the basis of feeling well and not taking drugs), assayed monthly over a period of six months.
  • FIG. 14 shows the effect over time, on inflammatory gene expression in a single human subject, of the administration of an anti-inflammatory steroid, as assayed using the Inflammation Gene Expression Panel (which is incorporated in the Precision ProfileTM for Inflammatory Response shown in Table 10).
  • FIG. 15 shows the effect over time, via whole blood samples obtained from a human subject, administered a single dose of prednisone, on expression of 5 genes (of the Inflammation Gene Expression Panel (which is incorporated in the Precision ProfileTM for Inflammatory Response shown in Table 10).
  • FIG. 16 shows the effect over time, on inflammatory gene expression in a single human subject suffering from rheumatoid arthritis, of the administration of a TNF-inhibiting compound, but here the expression is shown in comparison to the cognate locus average previously determined (in connection with FIGS. 6 and 7 ) for the normal (i.e., undiagnosed, healthy) population.
  • FIG. 17A illustrates the consistency of inflammatory gene expression in a population.
  • FIG. 17B shows the normal distribution of index values obtained from an undiagnosed population.
  • FIG. 17C illustrates the use of the same index as FIG. 17B , where the inflammation median for a normal population has been set to zero and both normal and diseased subjects are plotted in standard deviation units relative to that median.
  • FIG. 18 plots, in a fashion similar to that of FIG. 17A , Gene Expression Profiles, for the same 7 loci as in FIG. 17A , two different (responder v. non-responder) 6-subject populations of rheumatoid arthritis patients.
  • FIG. 19 illustrates use of the inflammation index for assessment of a single subject suffering from rheumatoid arthritis, who has not responded well to traditional therapy with methotrexate.
  • FIG. 20 illustrates use of the inflammation index for assessment of three subjects suffering from rheumatoid arthritis, who have not responded well to traditional therapy with methotrexate.
  • FIG. 21 shows the inflammation index for an international group of subjects, suffering from rheumatoid arthritis, undergoing three separate treatment regimens
  • FIG. 22 shows the inflammation index for an international group of subjects, suffering from rheumatoid arthritis, undergoing three separate treatment regimens
  • FIG. 23 shows the inflammation index for an international group of subjects, suffering from rheumatoid arthritis, undergoing three separate treatment regimens.
  • FIG. 24 illustrates use of the inflammation index for assessment of a single subject suffering from inflammatory bowel disease.
  • FIG. 25 shows Gene Expression Profiles with respect to 24 loci (of the Inflammation Gene Expression Panel (which is incorporated in the Precision ProfileTM for Inflammatory Response shown in Table 10) for whole blood treated with Ibuprofen in vitro in relation to other non-steroidal anti-inflammatory drugs (NSAIDs).
  • NSAIDs non-steroidal anti-inflammatory drugs
  • FIG. 26 illustrates how the effects of two competing anti-inflammatory compounds can be compared objectively, quantitatively, precisely, and reproducibly.
  • FIG. 27 uses a novel bacterial Gene Expression Panel of 24 genes, developed to discriminate various bacterial conditions in a host biological system.
  • FIG. 28 shows differential expression for a single locus, IFNG, to LTA derived from three distinct sources: S. pyrogenes, B. subtilis , and S. aureus.
  • FIG. 29 shows the response after two hours of the Inflammation 48A and 48B loci respectively (discussed above in connection with FIGS. 6 and 7 respectively) in whole blood to administration of a Gram-positive and a Gram-negative organism.
  • FIG. 30 shows the response after two hours of the Inflammation 48A and 48B loci respectively (discussed above in connection with FIGS. 6 and 7 respectively) in whole blood to administration of a Gram-positive and a Gram-negative organism.
  • FIG. 31 shows the response after six hours of the Inflammation 48A and 48B loci respectively (discussed above in connection with FIGS. 6 and 7 respectively) in whole blood to administration of a Gram-positive and a Gram-negative organism.
  • FIG. 32 shows the response after six hours of the Inflammation 48A and 48B loci respectively (discussed above in connection with FIGS. 6 and 7 respectively) in whole blood to administration of a Gram-positive and a Gram-negative organism.
  • FIG. 33 compares the gene expression response induced by E. coli and by an organism-free E. coli filtrate.
  • FIG. 34 is similar to FIG. 33 , but compared responses are to stimuli from E. coli filtrate alone and from E. coli filtrate to which has been added polymyxin B.
  • FIG. 35 illustrates the gene expression responses induced by S. aureus at 2, 6, and 24 hours after administration.
  • FIG. 36 illustrates the comparison of the gene expression induced by E. coli and S. aureus under various concentrations and times.
  • FIG. 37 illustrates the comparison of the gene expression induced by E. coli and S. aureus under various concentrations and times.
  • FIG. 38 illustrates the comparison of the gene expression induced by E. coli and S. aureus under various concentrations and times.
  • FIG. 39 illustrates the comparison of the gene expression induced by E. coli and S. aureus under various concentrations and times.
  • FIG. 40 illustrates the comparison of the gene expression induced by E. coli and S. aureus under various concentrations and times.
  • FIG. 41 illustrates the comparison of the gene expression induced by E. coli and S. aureus under various concentrations and times.
  • FIG. 42 illustrates application of a statistical T-test to identify potential members of a signature gene expression panel that is capable of distinguishing between normal subjects and subjects suffering from unstable rheumatoid arthritis.
  • FIG. 43 illustrates for a panel of 17 genes, the expression levels for 8 patients presumed to have bacteremia.
  • FIG. 44 illustrates application of a statistical T-test to identify potential members of a signature gene expression panel that is capable of distinguishing between normal subjects and subjects suffering from bacteremia
  • FIG. 45 illustrates application of an algorithm (shown in the figure), providing an index pertinent to rheumatoid arthritis (RA) as applied respectively to normal subjects, RA patients, and bacteremia patients.
  • RA rheumatoid arthritis
  • FIG. 46 illustrates application of an algorithm (shown in the figure), providing an index pertinent to bacteremia as applied respectively to normal subjects, rheumatoid arthritis patients, and bacteremia patients.
  • FIG. 47 illustrates, for a panel of 47 genes selected genes from Table 1, the expression levels for a patient suffering from multiple sclerosis on dates May 22, 2002 (no treatment), May 28, 2002 (after 5 mg prednisone given on May 22), and Jul. 15, 2002 (after 100 mg prednisone given on May 28, tapering to 5 mg within one week).
  • FIG. 48 shows a scatter plot of a three-gene model useful for discriminating MS subjects generated by Latent Class Modeling analysis using ITGAM with MMP9 and ITGA4.
  • FIG. 49 shows a scatter plot of an alternative three-gene model useful for discriminating MS subjects using ITGAM with CD4 and MMP9.
  • FIG. 50 shows a scatter plot of the same alternative three-gene model of FIG. 49 useful for discriminating MS subjects using ITGAM with MMP9 and CD4 but now displaying only washed out subjects relative to normals.
  • FIG. 51 shows a scatter plot of a four-gene model useful for discriminating MS subjects using ITGAM with ITGA4, MMP9 and CALCA.
  • FIG. 52 shows a scatter plot of a five-gene model useful for discriminating MS subjects using ITGAM with ITGA4, NFKB1B, MMP9 and CALCA.
  • FIG. 53 shows another five-gene model useful for discriminating MS subjects using ITGAM with ITGA4, NFKB1B, MMP9 and CXCR3 replacing CALCA.
  • FIG. 54 show a shows a four-gene model useful for discriminating MS subjects using ITGAL, CASP9, HLADRA and TGFBR2.
  • FIG. 55 show a shows a two-gene model useful for discriminating MS subjects using CASP9 and HLADRA.
  • FIG. 56 show a shows a two-gene model useful for discriminating MS subjects using ITGAL and HLADRA.
  • FIG. 57 show a shows a three-gene model useful for discriminating MS subjects using ITGAL, CASP9, and HLADRA.
  • Algorithm is a set of rules for describing a biological condition.
  • the rule set may be defined exclusively algebraically but may also include alternative or multiple decision points requiring domain-specific knowledge, expert interpretation or other clinical indicators.
  • composition or a “stimulus”, as those terms are defined herein, or a combination of a composition and a stimulus.
  • Amplification in the context of a quantitative RT-PCR assay is a function of the number of DNA replications that are tracked to provide a quantitative determination of its concentration. “Amplification” here refers to a degree of sensitivity and specificity of a quantitative assay technique. Accordingly, amplification provides a measurement of concentrations of constituents that is evaluated under conditions wherein the efficiency of amplification and therefore the degree of sensitivity and reproducibility for measuring all constituents is substantially similar.
  • a “baseline profile data set” is a set of values associated with constituents of a Gene Expression Panel resulting from evaluation of a biological sample (or population or set of samples) under a desired biological condition that is used for mathematically normative purposes.
  • the desired biological condition may be, for example, the condition of a subject (or population or set of subjects) before exposure to an agent or in the presence of an untreated disease or in the absence of a disease.
  • the desired biological condition may be health of a subject or a population or set of subjects.
  • the desired biological condition may be that associated with a population or set of subjects selected on the basis of at least one of age group, gender, ethnicity, geographic location, nutritional history, medical condition, clinical indicator, medication, physical activity, body mass, and environmental exposure.
  • a “biological condition” of a subject is the condition of the subject in a pertinent realm that is under observation, and such realm may include any aspect of the subject capable of being monitored for change in condition, such as health, disease including cancer; autoimmune condition; trauma; aging; infection; tissue degeneration; developmental steps; physical fitness; obesity, and mood.
  • a condition in this context may be chronic or acute or simply transient.
  • a targeted biological condition may be manifest throughout the organism or population of cells or may be restricted to a specific organ (such as skin, heart, eye or blood), but in either case, the condition may be monitored directly by a sample of the affected population of cells or indirectly by a sample derived elsewhere from the subject.
  • the term “biological condition” includes a “physiological condition”.
  • Body fluid of a subject includes blood, urine, spinal fluid, lymph, mucosal secretions, prostatic fluid, semen, haemolymph or any other body fluid known in the art for a subject.
  • “Calibrated profile data set” is a function of a member of a first profile data set and a corresponding member of a baseline profile data set for a given constituent in a panel.
  • a “clinical indicator” is any physiological datum used alone or in conjunction with other data in evaluating the physiological condition of a collection of cells or of an organism. This term includes pre-clinical indicators.
  • a “composition” includes a chemical compound, a nutraceutical, a pharmaceutical, a homeopathic formulation, an allopathic formulation, a naturopathic formulation, a combination of compounds, a toxin, a food, a food supplement, a mineral, and a complex mixture of substances, in any physical state or in a combination of physical states.
  • a profile data set from a sample includes determining a set of values associated with constituents of a Gene Expression Panel either (i) by direct measurement of such constituents in a biological sample or (ii) by measurement of such constituents in a second biological sample that has been exposed to the original sample or to matter derived from the original sample.
  • a “Gene Expression Panel” is an experimentally verified set of constituents, each constituent being a distinct expressed product of a gene, whether RNA or protein, wherein constituents of the set are selected so that their measurement provides a measurement of a targeted biological condition.
  • a “Gene Expression Profile” is a set of values associated with constituents of a Gene Expression Panel resulting from evaluation of a biological sample (or population or set of samples).
  • a “Gene Expression Profile Inflammatory Index” is the value of an index function that provides a mapping from an instance of a Gene Expression Profile into a single-valued measure of inflammatory condition.
  • “Inflammatory state” is used to indicate the relative biological condition of a subject resulting from inflammation, or characterizing the degree of inflammation
  • a “large number” of data sets based on a common panel of genes is a number of data sets sufficiently large to permit a statistically significant conclusion to be drawn with respect to an instance of a data set based on the same panel.
  • a “normative” condition of a subject to whom a composition is to be administered means the condition of a subject before administration, even if the subject happens to be suffering from a disease.
  • a “panel” of genes is a set of genes including at least two constituents.
  • a “set” or “population” of samples or subjects refers to a defined or selected group of samples or subjects wherein there is an underlying commonality or relationship between the members included in the set or population of samples or subjects.
  • a “subject” is a cell, tissue, or organism, human or non-human, whether in vivo, ex vivo or in vitro, under observation.
  • evaluating the biological condition of a subject based on a sample from the subject we include using blood or other tissue sample from a human subject to evaluate the human subject's condition; but we also include, for example, using a blood sample itself as the subject to evaluate, for example, the effect of therapy or an agent upon the sample.
  • a “stimulus” includes (i) a monitored physical interaction with a subject, for example ultraviolet A or B, or light therapy for seasonal affective disorder, or treatment of psoriasis with psoralen or treatment of melanoma with embedded radioactive seeds, other radiation exposure, and (ii) any monitored physical, mental, emotional, or spiritual activity or inactivity of a subject.
  • Gene Expression Panels may be used for measurement of therapeutic efficacy of natural or synthetic compositions or stimuli that may be formulated individually or in combinations or mixtures for a range of targeted biological conditions; prediction of toxicological effects and dose effectiveness of a composition or mixture of compositions for an individual or for a population or set of individuals or for a population of cells; determination of how two or more different agents administered in a single treatment might interact so as to detect any of synergistic, additive, negative, neutral or toxic activity; performing pre-clinical and clinical trials by providing new criteria for pre-selecting subjects according to informative profile data sets for revealing disease status; and conducting preliminary dosage studies for these patients prior to conducting phase 1 or 2 trials.
  • These Gene Expression Panels may be employed with respect to samples derived from subjects in order to evaluate their biological condition.
  • the present invention provides Gene Expression Panels for the evaluation of multiple sclerosis and inflammatory condition related to multiple sclerosis.
  • Gene Expression Profiles described herein also provided the evaluation of the effect of one or more agents for the treatment of multiple sclerosis and inflammatory condition related to multiple sclerosis.
  • a Precision ProfileTM for Multiple Sclerosis or Inflammatory Conditions Related to Multiple Sclerosis includes one or more genes, e.g., constituents, listed in 1-9.
  • Each gene of the Precision ProfileTM for Multiple Sclerosis or Inflammatory Conditions Related to Multiple Sclerosis is referred to herein as a multiple sclerosis associated gene or a multiple sclerosis associated constituent.
  • the evaluation or characterization of multiple sclerosis is defined to be diagnosing multiple sclerosis, assessing the risk of developing multiple sclerosis or assessing the prognosis of a subject with multiple sclerosis.
  • the evaluation or characterization of an agent for treatment of multiple sclerosis includes identifying agents suitable for the treatment of multiple sclerosis.
  • the agents can be compounds known to treat multiple sclerosis or compounds that have not been shown to treat multiple sclerosis.
  • Multiple sclerosis and conditions related to multiple sclerosis is evaluated by determining the level of expression (e.g., a quantitative measure) of one or more multiple sclerosis genes.
  • the level of expression is determined by any means known in the art, such as for example quantitative PCR.
  • the measurement is obtained under conditions that are substantially repeatable.
  • the qualitative measure of the constituent is compared to a baseline level (e.g. baseline profile set).
  • a baseline level is a level of expression of the constituent in one or more subjects known not to be suffering from multiple sclerosis (e.g., normal, healthy individual(s)).
  • the baseline level is derived from one or more subjects known to be suffering from multiple sclerosis.
  • the baseline level is derived from the same subject from which the first measure is derived.
  • the baseline is taken from a subject at different time periods during a course of treatment.
  • Such methods allow for the evaluation of a particular treatment for a selected individual. Comparison can be performed on test (e.g., patient) and reference samples (e.g., baseline) measured concurrently or at temporally distinct times.
  • test e.g., patient
  • reference samples e.g., baseline
  • An example of the latter is the use of compiled expression information, e.g., a gene expression database, which assembles information about expression levels of multiple sclerosis genes.
  • a change in the expression pattern in the patient-derived sample of a multiple sclerosis gene compared to the normal baseline level indicates that the subject is suffering from or is at risk of developing multiple sclerosis.
  • a similar level compared to the normal control level in the patient-derived sample of a multiple sclerosis gene indicates that the subject is not suffering from or is at risk of developing multiple sclerosis.
  • a similarity in the expression pattern in the patient-derived sample of a multiple sclerosis gene compared to the multiple sclerosis baseline level indicates that the subject is suffering from or is at risk of developing multiple sclerosis.
  • Expression of an effective amount of a multiple sclerosis gene also allows for the course of treatment of multiple sclerosis to be monitored.
  • a biological sample is provided from a subject undergoing treatment, e.g., if desired, biological samples are obtained from the subject at various time points before, during, or after treatment.
  • Expression of an effective amount of a multiple sclerosis gene is then determined and compared to baseline profile.
  • the baseline profile may be taken or derived from one or more individuals who have been exposed to the treatment.
  • the baseline level may be taken or derived from one or more individuals who have not been exposed to the treatment.
  • samples may be collected from subjects who have received initial treatment for multiple sclerosis and subsequent treatment for multiple sclerosis to monitor the progress of the treatment.
  • the Precision ProfileTM for Multiple Sclerosis and Inflammatory Conditions Related to Multiple Sclerosis allows for a putative therapeutic or prophylactic to be tested from a selected subject in order to determine if the agent is a suitable for treating or preventing multiple sclerosis in the subject. Additionally, other genes known to be associated with toxicity may be used.
  • suitable for treatment is meant determining whether the agent will be efficacious, not efficacious, or toxic for a particular individual.
  • toxic it is meant that the manifestations of one or more adverse effects of a drug when administered therapeutically. For example, a drug is toxic when it disrupts one or more normal physiological pathways.
  • test sample from the subject is exposed to a candidate therapeutic agent, and the expression of one or more of multiple sclerosis genes is determined.
  • a subject sample is incubated in the presence of a candidate agent and the pattern of multiple sclerosis gene expression in the test sample is measured and compared to a baseline profile, e.g., a multiple sclerosis baseline profile or a non-multiple sclerosis baseline profile or an index value.
  • the test agent can be any compound or composition.
  • the test agent is a compound known to be useful in the treatment of multiple sclerosis.
  • the test agent is a compound that has not previously been used to treat multiple sclerosis.
  • the reference sample e.g., baseline is from a subject that does not have multiple sclerosis a similarity in the pattern of expression of multiple sclerosis genes in the test sample compared to the reference sample indicates that the treatment is efficacious. Whereas a change in the pattern of expression of multiple sclerosis genes in the test sample compared to the reference sample indicates a less favorable clinical outcome or prognosis.
  • Efficacious is meant that the treatment leads to a decrease of a sign or symptom of multiple sclerosis in the subject or a change in the pattern of expression of a multiple sclerosis gene in such that the gene expression pattern has an increase in similarity to that of a normal baseline pattern.
  • Assessment of multiple sclerosis is made using standard clinical protocols. Efficacy is determined in association with any known method for diagnosing or treating multiple sclerosis.
  • Agents that are toxic for a specific subject are identified by exposing a test sample from the subject to a candidate agent, and the expression of one or more of multiple sclerosis genes is determined.
  • a subject sample is incubated in the presence of a candidate agent and the pattern of multiple sclerosis gene expression in the test sample is measured and compared to a baseline profile, e.g., a multiple sclerosis baseline profile or a non-multiple sclerosis baseline profile or an index value.
  • the test agent can be any compound or composition.
  • the test agent is a compound known to be useful in the treatment of multiple sclerosis.
  • the test agent is a compound that has not previously been used to treat multiple sclerosis.
  • the reference sample e.g., baseline is from a subject in whom the candidate agent is not toxic
  • a similarity in the pattern of expression of multiple sclerosis genes in the test sample compared to the reference sample indicates that the candidate agent is not toxic for the particular subject.
  • a change in the pattern of expression of multiple sclerosis genes in the test sample compared to the reference sample indicates that the candidate agent is toxic.
  • a Gene Expression Panel (Precision ProfileTM) is selected in a manner so that quantitative measurement of RNA or protein constituents in the Panel constitutes a measurement of a biological condition of a subject.
  • a calibrated profile data set is employed. Each member of the calibrated profile data set is a function of (i) a measure of a distinct constituent of a Gene Expression Panel (Precision ProfileTM) and (ii) a baseline quantity.
  • a degree of repeatability of measurement of better than twenty percent may be used as providing measurement conditions that are “substantially repeatable”.
  • expression levels for a constituent in a Gene Expression Panel may be meaningfully compared from sample to sample.
  • the criterion of repeatability means that all measurements for this constituent, if skewed, will nevertheless be skewed systematically, and therefore measurements of expression level of the constituent may be compared meaningfully. In this fashion valuable information may be obtained and compared concerning expression of the constituent under varied circumstances.
  • a second criterion also be satisfied, namely that quantitative measurement of constituents is performed under conditions wherein efficiencies of amplification for all constituents are substantially similar as defined herein.
  • measurement of the expression level of one constituent may be meaningfully compared with measurement of the expression level of another constituent in a given sample and from sample to sample.
  • Additional embodiments relate to the use of an index or algorithm resulting from quantitative measurement of constituents, and optionally in addition, derived from either expert analysis or computational biology (a) in the analysis of complex data sets; (b) to control or normalize the influence of uninformative or otherwise minor variances in gene expression values between samples or subjects; (c) to simplify the characterization of a complex data set for comparison to other complex data sets, databases or indices or algorithms derived from complex data sets; (d) to monitor a biological condition of a subject; (e) for measurement of therapeutic efficacy of natural or synthetic compositions or stimuli that may be formulated individually or in combinations or mixtures for a range of targeted biological conditions; (f) for predictions of toxicological effects and dose effectiveness of a composition or mixture of compositions for an individual or for a population or set of individuals or for a population of cells; (g) for determination of how two or more different agents administered in a single treatment might interact so as to detect any of synergistic, additive, negative, neutral of toxic activity (h) for performing pre-clin
  • Gene expression profiling and the use of index characterization for a particular condition or agent or both may be used to reduce the cost of Phase 3 clinical trials and may be used beyond Phase 3 trials; labeling for approved drugs; selection of suitable medication in a class of medications for a particular patient that is directed to their unique physiology; diagnosing or determining a prognosis of a medical condition or an infection which may precede onset of symptoms or alternatively diagnosing adverse side effects associated with administration of a therapeutic agent; managing the health care of a patient; and quality control for different batches of an agent or a mixture of agents.
  • the methods disclosed here may be applied to cells of humans, mammals or other organisms without the need for undue experimentation by one of ordinary skill in the art because all cells transcribe RNA and it is known in the art how to extract RNA from all types of cells.
  • a subject can include those who have not been previously diagnosed as having multiple sclerosis or an inflammatory condition related to multiple sclerosis. Alternatively, a subject can also include those who have already been diagnosed as having multiple sclerosis or an inflammatory condition related to multiple sclerosis. Diagnosis of multiple sclerosis is made for example, by clinical data (e.g., episodes of neurologic symptoms characteristic of MS and abnormalities upon physical examination), magnetic resonance imaging of the brain and spine to identify lesions and plaques, testing of cerebral spinal fluid for oligoclonal bands, and measurements of antibodies against myelin proteins (e.g., myelin oligodendrocyte glycoprotein (MOG) and myelin basic protein (MBP).
  • MEG myelin oligodendrocyte glycoprotein
  • MBP myelin basic protein
  • the subject has been previously treated with therapeutic agents, or with other therapies and treatment regimens for multiple sclerosis or an inflammatory condition related to multiple sclerosis.
  • a subject can also include those who are suffering from, or at risk of developing multiple sclerosis or an inflammatory condition related to multiple sclerosis, such as those who exhibit known risk factors for multiple sclerosis or an inflammatory condition related to multiple sclerosis.
  • Known risk factors for multiple sclerosis include but are not limited to viral infection, decrease exposure to sunlight, vitamin-D deficiency, chronic infection with spirochetal bacteria and/or Chlamydophila pneumonia, exposure to Epstein-Barr virus, severe stress, and smoking.
  • a subject can include those who are candidates for anti-TNF therapy.
  • Tables 1, 2, 3, 4, 5, 6, 7, 8, or 9 listed below include relevant genes which may be selected for a given Gene Expression Panel, such as the Gene Expression Panels demonstrated herein to be useful in the evaluation of multiple sclerosis and inflammatory condition related to multiple sclerosis.
  • Tables 1-2 were derived from a study of the gene expression patterns described in Example 12 below.
  • Tables 3 and 5-9 were derived from a study of gene expression patterns described in Example 13 below.
  • Table 4 is a panel of 104 genes whose expression is associated with Multiple Sclerosis or Inflammatory Conditions related to Multiple Sclerosis, referred to herein as the Precision ProfileTM for Multiple Sclerosis and Inflammatory Genes Related to Multiple Sclerosis.
  • Table 5 shows a ranking of p-values (from most to least significant) of a subset of genes from Table 4.
  • Table 6 describes 2-gene model based on genes from the Precision ProfileTM for Multiple sclerosis derived from latent class modeling of the subjects from this study to distinguish between subjects suffering from multiple sclerosis and normal subjects.
  • Two gene models capable of correctly classifying multiple sclerosis-afflicted and/or normal subjects with at least 75% accuracy are indicated.
  • 2-gene model, ITGAL and HLADRA correctly classifies multiple sclerosis-afflicted subjects with 85.4% accuracy, and normal subjects with 82.9% accuracy.
  • the 2-gene model, CASP9 and HLADRA correctly classifies multiple sclerosis-afflicted subjects with 78.5% accuracy, and normal subjects with 84.2% accuracy.
  • Table 7 describes 3-gene models based on genes from the Precision ProfileTM for Multiple Sclerosis, capable of correctly classifying multiple sclerosis-afflicted and/or normal subjects with at least 75% accuracy are indicated.
  • the three-gene model, ITGAL, HLADRA, and CASP9 correctly classifies multiple sclerosis-afflicted subjects with 85.4% accuracy, and normal subjects with 86.8% accuracy.
  • Table 8 describes a 4-gene model based on genes from the Precision ProfileTM for Multiple Sclerosis, capable of correctly classifying multiple sclerosis-afflicted and/or normal subjects with at least 75% accuracy are indicated.
  • the 4-gene model CASP9, HLADRA, ITGAL, and CCR3, correctly classifies multiple sclerosis-afflicted subjects with 85.4% accuracy, and normal subjects with 83.6% accuracy.
  • Table 9 describes a 5-gene model based on genes from the Precision ProfileTM for Multiple Sclerosis, capable of correctly classifying multiple sclerosis-afflicted and/or normal subjects with at least 75% accuracy are indicated.
  • the 4-gene model CASP9, HLADRA, ITGAL, CCR3, and TGFBR2 correctly classifies multiple sclerosis-afflicted subjects with 86.9% accuracy, and normal subjects with 84.2% accuracy.
  • Table 10 is a panel of 96 genes whose expression is associated with Inflammation referred to herein as the Precision ProfileTM for Inflammatory Response.
  • panels may be constructed and experimentally verified by one of ordinary skill in the art in accordance with the principles articulated in the present application.
  • intra-assay variability This is a measure called “intra-assay variability”. Assays have also been conducted on different occasions using the same sample material. This is a measure of “inter-assay variability”.
  • the average coefficient of variation of intra-assay variability or inter-assay variability is less than 20%, more preferably less than 10%, more preferably less than 5%, more preferably less than 4%, more preferably less than 3%, more preferably less than 2%, and even more preferably less than 1%.
  • RNA is extracted from a sample such as any tissue, body fluid, cell, or culture medium in which a population of cells of a subject might be growing. For example, cells may be lysed and RNA eluted in a suitable solution in which to conduct a DNAse reaction. Subsequent to RNA extraction, first strand synthesis may be performed using a reverse transcriptase.
  • Gene amplification more specifically quantitative PCR assays, can then be conducted and the gene of interest calibrated against an internal marker such as 18S rRNA (Hirayama et al., Blood 92, 1998: 46-52). Any other endogenous marker can be used, such as 28S-25S rRNA and 5S rRNA. Samples are measured in multiple replicates, for example, 3 replicates.
  • quantitative PCR is performed using amplification, reporting agents and instruments such as those supplied commercially by Applied Biosystems (Foster City, Calif.). Given a defined efficiency of amplification of target transcripts, the point (e.g., cycle number) that signal from amplified target template is detectable may be directly related to the amount of specific message transcript in the measured sample.
  • quantifiable signals such as fluorescence, enzyme activity, disintegrations per minute, absorbance, etc.
  • a known concentration of target templates e.g., a reference standard curve
  • a standard with limited variability can be used to quantify the number of target templates in an unknown sample.
  • quantitative gene expression techniques may utilize amplification of the target transcript.
  • quantitation of the reporter signal for an internal marker generated by the exponential increase of amplified product may also be used.
  • Amplification of the target template may be accomplished by isothermic gene amplification strategies or by gene amplification by thermal cycling such as PCR.
  • Amplification efficiencies are regarded as being “substantially similar”, for the purposes of this description and the following claims, if they differ by no more than approximately 10%, preferably by less than approximately 5%, more preferably by less than approximately 3%, and more preferably by less than approximately 1%.
  • Measurement conditions are regarded as being “substantially repeatable, for the purposes of this description and the following claims, if they differ by no more than approximately +/ ⁇ 10% coefficient of variation (CV), preferably by less than approximately +/ ⁇ 5% CV, more preferably +/ ⁇ 2% CV.
  • primer-probe design can be enhanced using computer techniques known in the art, and notwithstanding common practice, it has been found that experimental validation is still useful. Moreover, in the course of experimental validation, the selected primer-probe combination is associated with a set of features:
  • the reverse primer should be complementary to the coding DNA strand.
  • the primer should be located across an intron-exon junction, with not more than four bases of the three-prime end of the reverse primer complementary to the proximal exon. (If more than four bases are complementary, then it would tend to competitively amplify genomic DNA.)
  • the primer probe set should amplify cDNA of less than 110 bases in length and should not amplify, or generate fluorescent signal from, genomic DNA or transcripts or cDNA from related but biologically irrelevant loci.
  • a suitable target of the selected primer probe is first strand cDNA, which in one embodiment may be prepared from whole blood as follows:
  • Human blood is obtained by venipuncture and prepared for assay by separating samples for baseline, no exogenous stimulus, and pro-cancer stimulus with sufficient volume for at least three time points.
  • Typical pro-cancer stimuli include for example, ionizing radiation, free radicals or DNA damaging agents, and may be used individually or in combination.
  • the aliquots of heparinized, whole blood are mixed with additional test therapeutic compounds and held at 37° C. in an atmosphere of 5% CO 2 for 30 minutes. Stimulus is added at varying concentrations, mixed and held loosely capped at 37° C. for the prescribed timecourse.
  • cells are lysed and RNA extracted by various standard means.
  • RNA is preferentially obtained from the nucleic acid mix using a variety of standard procedures (or RNA Isolation Strategies, pp. 55-104, in RNA Methodologies, A laboratory guide for isolation and characterization, 2nd edition, 1998, Robert E. Farrell, Jr., Ed., Academic Press), in the present using a filter-based RNA isolation system from Anibion (RNAqueousTM, Phenol-free Total RNA Isolation Kit, Catalog #1912, version 9908; Austin, Tex.).
  • the whole blood assay for Gene Expression Profiles determination was carried out as follows: Human whole blood was drawn into 10 mL Vacutainer tubes with Sodium Heparin. Blood samples were mixed by gently inverting tubes 4-5 times. The blood was used within 10-15 minutes of draw. In the experiments, blood was diluted 2-fold, i.e. per sample per time point, 0.6 mL whole blood +0.6 mL stimulus. The assay medium was prepared and the stimulus added as appropriate.
  • a quantity (0.6 mL) of whole blood was then added into each 12 ⁇ 75 mm polypropylene tube.
  • 0.6 mL of 2 ⁇ LPS from E. coli serotype 0127:B8, Sigma#L3880 or serotype 055, Sigma #L4005, 10 ng/mL, subject to change in different lots
  • 2 ⁇ LPS from E. coli serotype 0127:B8, Sigma#L3880 or serotype 055, Sigma #L4005, 10 ng/mL, subject to change in different lots
  • 0.6 mL assay medium was added to the “control” tubes.
  • the caps were closed tightly.
  • the tubes were inverted 2-3 times to mix samples. Caps were loosened to first stop and the tubes incubated at 37° C., 5% CO 2 for 6 hours.
  • samples were gently mixed to resuspend blood cells, and 0.15 mL was removed from each tube (using a micropipettor with barrier tip), and transferred to 0.15 mL of lysis buffer and mixed. Lysed samples were extracted using an ABI 6100 Nucleic Acid Prepstation following the manufacturer's recommended protocol.
  • RNA samples were then centrifuged for 5 min at 500 ⁇ g, ambient temperature (IEC centrifuge or equivalent, in microfuge tube adapters in swinging bucket), and as much serum from each tube was removed as possible and discarded.
  • Cell pellets were placed on ice; and RNA extracted as soon as possible using an Ambion RNAqueous kit.
  • RNAs are amplified using message specific primers or random primers.
  • the specific primers are synthesized from data obtained from public databases (e.g., Unigene, National Center for Biotechnology Information, National Library of Medicine, Bethesda, Md.), including information from genomic and cDNA libraries obtained from humans and other animals. Primers are chosen to preferentially amplify from specific RNAs obtained from the test or indicator samples (see, for example, RT PCR, Chapter 15 in RNA Methodologies, A laboratory guide for isolation and characterization, 2nd edition, 1998, Robert E. Farrell, Jr., Ed., Academic Press; or Chapter 22 pp. 143-151 , RNA isolation and characterization protocols , Methods in molecular biology, Volume 86, 1998, R.
  • Amplifications are carried out in either isothermic conditions or using a thermal cycler (for example, a ABI 9600 or 9700 or 7900 obtained from Applied Biosystems, Foster City, Calif.; see Nucleic acid detection methods, pp. 1-24, in Molecular methods for virus detection , D. L. Wiedbrauk and D. H., Farkas, Eds., 1995, Academic Press).
  • a thermal cycler for example, a ABI 9600 or 9700 or 7900 obtained from Applied Biosystems, Foster City, Calif.; see Nucleic acid detection methods, pp. 1-24, in Molecular methods for virus detection , D. L. Wiedbrauk and D. H., Farkas, Eds., 1995, Academic Press.
  • Amplified nucleic acids are detected using fluorescent-tagged detection oligonucleotide probes (see, for example, TaqmanTM PCR Reagent Kit, Protocol, part number 402823, Revision A, 1996, Applied Biosystems, Foster City Calif.) that are identified and synthesized from publicly known databases as described for the amplification primers.
  • amplified cDNA is detected and quantified using the ABI Prism 7900 Sequence Detection System obtained from Applied Biosystems (Foster City, Calif.).
  • Amounts of specific RNAs contained in the test sample or obtained from the indicator cell lines can be related to the relative quantity of fluorescence observed (see for example, Advances in quantitative PCR technology: 5′ nuclease assays, Y. S. Lie and C. J. Petropolus, Current Opinion in Biotechnology, 1998, 9:43-48, or Rapid thermal cycling and PCR kinetics, pp. 211-229, chapter 14 in PCR applications: protocols for functional genomics, M. A. Innis, D. H. Gelfand and J. J. Sninsky, Eds., 1999, Academic Press).
  • Kit Components 10 ⁇ TaqMan RT Buffer, 25 mM Magnesium chloride, deoxyNTPs mixture, Random Hexamers, RNase Inhibitor, MultiScribe Reverse Transcriptase (50 U/mL) (2) RNase/DNase free water (DEPC Treated Water from Ambion (P/N 9915G), or equivalent).
  • RNA samples from ⁇ 80° C. freezer and thaw at room temperature and then place immediately on ice.
  • reaction e.g. 10 samples ( ⁇ L) 10X RT Buffer 10.0 110.0 25 mM MgCl 2 22.0 242.0 dNTPs 20.0 220.0 Random Hexamers 5.0 55.0 RNAse Inhibitor 2.0 22.0 Reverse Transcriptase 2.5 27.5 Water 18.5 203.5 Total: 80.0 880.0 (80 ⁇ L per sample)
  • RNA sample to a total volume of 20 ⁇ L in a 1.5 mL microcentrifuge tube (for example, for THP-1 RNA, remove 10 ⁇ L RNA and dilute to 20 ⁇ L with RNase/DNase free water, for whole blood RNA use 20 ⁇ L total RNA) and add 80 ⁇ L RT reaction mix from step 5, 2, 3. Mix by pipetting up and down.
  • a 1.5 mL microcentrifuge tube for example, for THP-1 RNA, remove 10 ⁇ L RNA and dilute to 20 ⁇ L with RNase/DNase free water, for whole blood RNA use 20 ⁇ L total RNA
  • PCR QC should be run on all RT samples using 18S and ⁇ -actin.
  • the use of the primer probe with the first strand cDNA as described above to permit measurement of constituents of a Gene Expression Panel is performed using a QPCR assay on Cepheid SmartCycler® and GeneXpert® Instruments as follows:
  • SmartMix TM-HM lyophilized Master Mix 1 bead 20X 18S Primer/Probe Mix 2.5 ⁇ L 20X Target Gene 1 Primer/Probe Mix 2.5 ⁇ L 20X Target Gene 2 Primer/Probe Mix 2.5 ⁇ L 20X Target Gene 3 Primer/Probe Mix 2.5 ⁇ L Tris Buffer, pH 9.0 2.5 ⁇ L Sterile Water 34.5 ⁇ L Total 47 ⁇ L
  • SmartMix TM-HM lyophilized Master Mix 1 bead SmartBead TM containing four primer/probe sets 1 bead Tris Buffer, pH 9.0 2.5 ⁇ L Sterile Water 44.5 ⁇ L Total 47 ⁇ L
  • any tissue, body fluid, or cell(s) may be used for ex vivo assessment of a biological condition affected by an agent.
  • Methods herein may also be applied using proteins where sensitive quantitative techniques, such as an Enzyme Linked ImmunoSorbent Assay (ELISA) or mass spectroscopy, are available and well-known in the art for measuring the amount of a protein constituent. (see WO 98/24935 herein incorporated by reference).
  • ELISA Enzyme Linked ImmunoSorbent Assay
  • mass spectroscopy mass spectroscopy
  • the analyses of samples from single individuals and from large groups of individuals provide a library of profile data sets relating to a particular panel or series of panels. These profile data sets may be stored as records in a library for use as baseline profile data sets. As the term “baseline” suggests, the stored baseline profile data sets serve as comparators for providing a calibrated profile data set that is informative about a biological condition or agent. Baseline profile data sets may be stored in libraries and classified in a number of cross-referential ways. One form of classification may rely on the characteristics of the panels from which the data sets are derived. Another form of classification may be by particular biological condition, e.g., multiple sclerosis. The concept of biological condition encompasses any state in which a cell or population of cells may be found at any one time.
  • This state may reflect geography of samples, sex of subjects or any other discriminator. Some of the discriminators may overlap.
  • the libraries may also be accessed for records associated with a single subject or particular clinical trial.
  • the classification of baseline profile data sets may further be annotated with medical information about a particular subject, a medical condition, and/or a particular agent.
  • the choice of a baseline profile data set for creating a calibrated profile data set is related to the biological condition to be evaluated, monitored, or predicted, as well as, the intended use of the calibrated panel, e.g., as to monitor drug development, quality control or other uses. It may be desirable to access baseline profile data sets from the same subject for whom a first profile data set is obtained or from different subject at varying times, exposures to stimuli, drugs or complex compounds; or may be derived from like or dissimilar populations or sets of subjects.
  • the baseline profile data set may be normal, healthy baseline.
  • the profile data set may arise from the same subject for which the first data set is obtained, where the sample is taken at a separate or similar time, a different or similar site or in a different or similar biological condition.
  • a sample may be taken before stimulation or after stimulation with an exogenous compound or substance, such as before or after therapeutic treatment.
  • the profile data set obtained from the unstimulated sample may serve as a baseline profile data set for the sample taken after stimulation.
  • the baseline data set may also be derived from a library containing profile data sets of a population or set of subjects having some defining characteristic or biological condition.
  • the baseline profile data set may also correspond to some ex vivo or in vitro properties associated with an in vitro cell culture.
  • the resultant calibrated profile data sets may then be stored as a record in a database or library along with or separate from the baseline profile data base and optionally the first profile data set although the first profile data set would normally become incorporated into a baseline profile data set under suitable classification criteria.
  • the remarkable consistency of Gene Expression Profiles associated with a given biological condition makes it valuable to store profile data, which can be used, among other things for normative reference purposes.
  • the normative reference can serve to indicate the degree to which a subject conforms to a given biological condition (healthy or diseased) and, alternatively or in addition, to provide a target for clinical intervention.
  • Selected baseline profile data sets may be also be used as a standard by which to judge manufacturing lots in terms of efficacy, toxicity, etc.
  • the baseline data set may correspond to Gene Expression Profiles taken before administration of the agent.
  • the baseline data set may correspond with a gold standard for that product.
  • any suitable normalization techniques may be employed. For example, an average baseline profile data set is obtained from authentic material of a naturally grown herbal nutraceutical and compared over time and over different lots in order to demonstrate consistency, or lack of consistency, in lots of compounds prepared for release.
  • an indicator cell line treated with an agent can in many cases provide calibrated profile data sets comparable to those obtained from in vivo or ex vivo populations of cells.
  • administering a sample from a subject onto indicator cells can provide informative calibrated profile data sets with respect to the biological condition of the subject including the health, disease states, therapeutic interventions, aging or exposure to environmental stimuli or toxins of the subject.
  • the calibrated profile data set may be expressed in a spreadsheet or represented graphically for example, in a bar chart or tabular form but may also be expressed in a three dimensional representation.
  • the function relating the baseline and profile data may be a ratio expressed as a logarithm.
  • the constituent may be itemized on the x-axis and the logarithmic scale may be on the y-axis.
  • Members of a calibrated data set may be expressed as a positive value representing a relative enhancement of gene expression or as a negative value representing a relative reduction in gene expression with respect to the baseline.
  • Patterns of this nature may be used to identify likely candidates for a drug trial, used alone or in combination with other clinical indicators to be diagnostic or prognostic with respect to a biological condition or may be used to guide the development of a pharmaceutical or nutraceutical through manufacture, testing and marketing.
  • the method also includes producing a calibrated profile data set for the panel, wherein each member of the calibrated profile data set is a function of a corresponding member of the first profile data set and a corresponding member of a baseline profile data set for the panel, and wherein the baseline profile data set is related to the multiple sclerosis or conditions related to multiple sclerosis to be evaluated, with the calibrated profile data set being a comparison between the first profile data set and the baseline profile data set, thereby providing evaluation of multiple sclerosis or conditions related to multiple sclerosis of the subject.
  • the function is a mathematical function and is other than a simple difference, including a second function of the ratio of the corresponding member of first profile data set to the corresponding member of the baseline profile data set, or a logarithmic function.
  • the first sample is obtained and the first profile data set quantified at a first location, and the calibrated profile data set is produced using a network to access a database stored on a digital storage medium in a second location, wherein the database may be updated to reflect the first profile data set quantified from the sample.
  • using a network may include accessing a global computer network.
  • the data is in a universal format, data handling may readily be done with a computer.
  • the data is organized so as to provide an output optionally corresponding to a graphical representation of a calibrated data set.
  • a distinct sample derived from a subject being at least one of RNA or protein may be denoted as PI.
  • the first profile data set derived from sample PI is denoted Mj, where Mj is a quantitative measure of a distinct RNA or protein constituent of PI.
  • the record Ri is a ratio of M and P and may be annotated with additional data on the subject relating to, for example, age, diet, ethnicity, gender, geographic location, medical disorder, mental disorder, medication, physical activity, body mass and environmental exposure.
  • data handling may further include accessing data from a second condition database which may contain additional medical data not presently held with the calibrated profile data sets. In this context, data access may be via a computer network.
  • the above described data storage on a computer may provide the information in a form that can be accessed by a user. Accordingly, the user may load the information onto a second access site including downloading the information. However, access may be restricted to users having a password or other security device so as to protect the medical records contained within.
  • a feature of this embodiment of the invention is the ability of a user to add new or annotated records to the data set so the records become part of the biological information.
  • the graphical representation of calibrated profile data sets pertaining to a product such as a drug provides an opportunity for standardizing a product by means of the calibrated profile, more particularly a signature profile.
  • the profile may be used as a feature with which to demonstrate relative efficacy, differences in mechanisms of actions, etc. compared to other drugs approved for similar or different uses.
  • the various embodiments of the invention may be also implemented as a computer program product for use with a computer system.
  • the product may include program code for deriving a first profile data set and for producing calibrated profiles.
  • Such implementation may include a series of computer instructions fixed either on a tangible medium, such as a computer readable medium (for example, a diskette, CD-ROM, ROM, or fixed disk), or transmittable to a computer system via a modem or other interface device, such as a communications adapter coupled to a network.
  • the network coupling may be for example, over optical or wired communications lines or via wireless techniques (for example, microwave, infrared or other transmission techniques) or some combination of these.
  • the series of computer instructions preferably embodies all or part of the functionality previously described herein with respect to the system.
  • Such computer instructions can be written in a number of programming languages for use with many computer architectures or operating systems. Furthermore, such instructions may be stored in any memory device, such as semiconductor, magnetic, optical or other memory devices, and may be transmitted using any communications technology, such as optical, infrared, microwave, or other transmission technologies. It is expected that such a computer program product may be distributed as a removable medium with accompanying printed or electronic documentation (for example, shrink wrapped software), preloaded with a computer system (for example, on system ROM or fixed disk), or distributed from a server or electronic bulletin board over a network (for example, the Internet or World Wide Web).
  • a computer system is further provided including derivative modules for deriving a first data set and a calibration profile data set.
  • the calibration profile data sets in graphical or tabular form, the associated databases, and the calculated index or derived algorithm, together with information extracted from the panels, the databases, the data sets or the indices or algorithms are commodities that can be sold together or separately for a variety of purposes as described in WO 01/25473.
  • a clinical indicator may be used to assess the multiple sclerosis or inflammatory conditions related to multiple sclerosis of the relevant set of subjects by interpreting the calibrated profile data set in the context of at least one other clinical indicator, wherein the at least one other clinical indicator is selected from the group consisting of blood chemistry, urinalysis, X-ray or other radiological or metabolic imaging technique, other chemical assays, and physical findings.
  • An index may be constructed using an index function that maps values in a Gene Expression Profile into a single value that is pertinent to the biological condition at hand.
  • the values in a Gene Expression Profile are the amounts of each constituent of the Gene Expression Panel that corresponds to the Gene Expression Profile. These constituent amounts form a profile data set, and the index function generates a single value—the index—from the members of the profile data set.
  • the index function may conveniently be constructed as a linear sum of terms, each term being what we call a “contribution function” of a member of the profile data set.
  • the contribution function may be a constant times a power of a member of the profile data set. So the index function would have the form
  • I is the index
  • Mi is the value of the member i of the profile data set
  • Ci is a constant
  • P(i) is a power to which Mi is raised, the sum being formed for all integral values of i up to the number of members in the data set.
  • the values C i and P(i) may be determined in a number of ways, so that the index I is informative of the pertinent biological condition.
  • One way is to apply statistical techniques, such as latent class modeling, to the profile data sets to correlate clinical data or experimentally derived data, or other data pertinent to the biological condition.
  • statistical techniques such as latent class modeling
  • Latent Gold® the software from Statistical Innovations, Belmont, Mass., called Latent Gold®. See the web pages at statisticalinnovations.com/lg/, which are hereby incorporated herein by reference.
  • the index function for inflammation may be constructed, for example, in a manner that a greater degree of inflammation (as determined by a profile data set for the Precision ProfileTM for Inflammatory Response shown in Table 10) correlates with a large value of the index function.
  • each P(i) may be +1 or —I, depending on whether the constituent increases or decreases with increasing inflammation.
  • braces around a constituent designate measurement of such constituent and the constituents are a subset of the Inflammation Gene Expression Panel (Precision ProfileTM for Inflammatory Response).
  • an index that characterizes a Gene Expression Profile can also be provided with a normative value of the index function used to create the index.
  • This normative value can be determined with respect to a relevant population or set of subjects or samples or to a relevant population of cells, so that the index may be interpreted in relation to the normative value.
  • the relevant population or set of subjects or samples, or relevant population of cells may have in common a property that is at least one of age range, gender, ethnicity, geographic location, nutritional history, medical condition, clinical indicator, medication, physical activity, body mass, and environmental exposure.
  • the index can be constructed, in relation to a normative Gene Expression Profile for a population or set of healthy subjects, in such a way that a reading of approximately 1 characterizes normative Gene Expression Profiles of healthy subjects.
  • the biological condition that is the subject of the index is inflammation; a reading of 11n this example thus corresponds to a Gene Expression Profile that matches the norm for healthy subjects.
  • a substantially higher reading then may identify a subject experiencing an inflammatory condition.
  • the use of 1 as identifying a normative value is only one possible choice; another logical choice is to use 0 as identifying the normative value.
  • Still another embodiment is a method of providing an index that is indicative of multiple sclerosis or inflammatory conditions related to multiple sclerosis of a subject based on a first sample from the subject, the first sample providing a source of RNAs, the method comprising deriving from the first sample a profile data set, the profile data set including a plurality of members, each member being a quantitative measure of the amount of a distinct RNA constituent in a panel of constituents selected so that measurement of the constituents is indicative of the presumptive signs of multiple sclerosis, the panel including at least two of the constituents of any of the Tables 1-10.
  • At least one measure from the profile data set is applied to an index function that provides a mapping from at least one measure of the profile data set into one measure of the presumptive signs of multiple sclerosis, so as to produce an index pertinent to the multiple sclerosis or inflammatory conditions related to multiple sclerosis of the subject.
  • M i and M j are values respectively of the member i and member j of the profile data set having N members, and C i and C ij are constants.
  • the index function is simply the constant C 0 .
  • the index function is a linear expression, in a form used for examples herein.
  • a quadratic expression that is constructed as a meaningful identifier of rheumatoid arthritis (RA) is the following:
  • C 0 serves to calibrate this expression to the biological population of interest (such as RA), that is characterized by inflammation.
  • the odds when the index value associated with a subject equals 0, the odds are 50:50 of the subject's being MS vs normal. More generally, the predicted odds of being MS is [exp(I i )], and therefore the predicted probability of being MS is [exp(I i )]/[1+exp((I i )]. Thus, when the index exceeds 0, the predicted probability that a subject is MS is higher than 0.5, and when it falls below 0, the predicted probability is less than 0.5.
  • the value of C 0 may be adjusted to reflect the prior probability of being in this population based on known exogenous risk factors for the subject.
  • C 0 is adjusted as a function of the subject's risk factors, where the subject has prior probability p i of being RA based on such risk factors, the adjustment is made by increasing (decreasing) the unadjusted C 0 value by adding to C 0 the natural logarithm of the ratio of the prior odds of being RA taking into account the risk factors to the overall prior odds of being RA without taking into account the risk factors.
  • Yet another embodiment provides a method of using an index for differentiating a type of pathogen within a class of pathogens of interest in a subject with multiple sclerosis or inflammatory conditions related to multiple sclerosis, based on at least one sample from the subject, the method comprising providing at least one index according to any of the above disclosed embodiments for the subject, comparing the at least one index to at least one normative value of the index, determined with respect to at least one relevant set of subjects to obtain at least one difference, and using the at least one difference between the at least one index and the at least one normative value for the index to differentiate the type of pathogen from the class of pathogen.
  • the invention also includes an MS-detection reagent, i.e., nucleic acids that specifically identify one or more multiple sclerosis or inflammatory condition related to multiple sclerosis nucleic acids (e.g., any gene listed in Tables 1-10; referred to herein as MS-associated genes) by having homologous nucleic acid sequences, such as oligonucleotide sequences, complementary to a portion of the MS-associated genes nucleic acids or antibodies to proteins encoded by the MS-associated genes nucleic acids packaged together in the form of a kit.
  • the oligonucleotides can be fragments of the MS-associated genes genes.
  • the oligonucleotides can be 200, 150, 100, 50, 25, 10 or less nucleotides in length.
  • the kit may contain in separate containers a nucleic acid or antibody (either already bound to a solid matrix or packaged separately with reagents for binding them to the matrix), control formulations (positive and/or negative), and/or a detectable label. Instructions (i.e., written, tape, VCR, CD-ROM, etc.) for carrying out the assay may be included in the kit.
  • the assay may for example be in the form of PCR, a Northern hybridization or a sandwich ELISA as known in the art.
  • MS-associated genes detection reagents can be immobilized on a solid matrix such as a porous strip to form at least one MS-associated genes detection site.
  • the measurement or detection region of the porous strip may include a plurality of sites containing a nucleic acid.
  • a test strip may also contain sites for negative and/or positive controls. Alternatively, control sites can be located on a separate strip from the test strip.
  • the different detection sites may contain different amounts of immobilized nucleic acids, i.e., a higher amount in the first detection site and lesser amounts in subsequent sites.
  • the number of sites displaying a detectable signal provides a quantitative indication of the amount of MS-associated genes present in the sample.
  • the detection sites may be configured in any suitably detectable shape and are typically in the shape of a bar or dot spanning the width of a test strip.
  • multiple sclerosis detection genes can be labeled (e.g., with one or more fluorescent dyes) and immobilized on lyophilized beads to form at least one multiple sclerosis gene detection site.
  • the beads may also contain sites for negative and/or positive controls.
  • the number of sites displaying a detectable signal provides a quantitative indication of the amount of multiple sclerosis genes present in the sample.
  • the kit contains a nucleic acid substrate array comprising one or more nucleic acid sequences.
  • the nucleic acids on the array specifically identify one or more nucleic acid sequences represented by MS-associated genes (e.g., any gene listed in Tables 1-10).
  • MS-associated genes e.g., any gene listed in Tables 1-10.
  • the expression of 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 40 or 50 or more of the sequences represented by MS-associated genes can be identified by virtue of binding to the array.
  • the substrate array can be on, i.e., a solid substrate, i.e., a “chip” as described in U.S. Pat. No. 5,744,305.
  • the substrate array can be a solution array, i.e., Luminex, Cyvera, Vitra and Quantum Dots' Mosaic.
  • nucleic acid probes i.e., oligonucleotides, aptamers, siRNAs, antisense oligonucleotides, against any of the MS-associated genes in Tables 1-10.
  • the index value or algorithm can be used to reduce a complex data set to a single index value that is informative with respect to the inflammatory state of a subject. This is illustrated in FIGS. 1A and 1B .
  • FIG. 1A is entitled Source Precision Inflammation Profile Tracking of A Subject Results in a Large, Complex Data Set.
  • the figure shows the results of assaying 24 genes from the Inflammation Gene Expression Panel (Precision ProfileTM for Inflammatory Response) on eight separate days during the course of optic neuritis in a single male subject.
  • FIG. 1B shows use of an Acute Inflammation Index.
  • the data displayed in FIG. 1A above is shown in this figure after calculation using an index function proportional to the following mathematical expression: (1/4 ⁇ IL1A ⁇ +1/4 ⁇ IL1B ⁇ +1/4 ⁇ TNF ⁇ +1/4 ⁇ INFG ⁇ 1/ ⁇ IL10 ⁇ ).
  • the inflammatory state of a subject reveals information about the past progress of the biological condition, future progress, response to treatment, etc.
  • the Acute Inflammation Index may be used to reveal such information about the biological condition of a subject. This is illustrated in FIG. 2 .
  • results of the assay for inflammatory gene expression for each day (shown for 24 genes in each row of FIG. 1A ) is displayed as an individual histogram after calculation. The index reveals clear trends in inflammatory status that may correlated with therapeutic intervention ( FIG. 2 ).
  • FIG. 2 is a graphical illustration of the acute inflammation index calculated at 9 different, significant clinical milestones from blood obtained from a single patient treated medically with for optic neuritis. Changes in the index values for the Acute Inflammation Index correlate strongly with the expected effects of therapeutic intervention.
  • Four clinical milestones have been identified on top of the Acute Inflammation Index in this figure including (1) prior to treatment with steroids, (2) treatment with IV solumedrol at 1 gram per day, (3) post-treatment with oral prednisone at 60 mg per day tapered to 10 mg per day and (4) post treatment.
  • the data set is the same as for FIG. 1 .
  • the index is proportional to 1/4 ⁇ IL1A ⁇ +1/4 ⁇ IL1B ⁇ +1/4 ⁇ TNF ⁇ +1/4 ⁇ INFG ⁇ 1/ ⁇ IL10 ⁇ .
  • the acute inflammation index falls rapidly with treatment with IV steroid, goes up during less efficacious treatment with oral prednisone and returns to the pre-treatment level after the steroids have been discontinued and metabolized completely.
  • the acute inflammation index may be used as a common reference value for therapeutic compounds or interventions without common mechanisms of action.
  • the compound that induces a gene response to a compound as indicated by the index, but fails to ameliorate a known biological conditions may be compared to a different compounds with varying effectiveness in treating the biological condition.
  • FIG. 3 shows the effects of single dose treatment with 800 mg of ibuprofen in a single donor as characterized by the Acute Inflammation Index.
  • the acute inflammation index falls immediately after taking the non-steroidal anti-inflammatory ibuprofen and returns to baseline after 48 hours.
  • FIG. 4 shows that the calculated acute inflammation index displayed graphically for five different conditions including (A) untreated whole blood; (B) whole blood treated in vitro with DMSO, an non-active carrier compound; (C) otherwise unstimulated whole blood treated in vitro with dexamethasone (0.08 ug/ml); (D) whole blood stimulated in vitro with lipopolysaccharide, a known pro-inflammatory compound, (LPS, 1 ng/ml) and (E) whole blood treated in vitro with LPS (1 ng/ml) and dexamethasone (0.08 ug/ml).
  • Dexamethasone is used as a prescription compound that is commonly used medically as an anti-inflammatory steroid compound.
  • the acute inflammation index is calculated from the experimentally determined gene expression levels of inflammation-related genes expressed in human whole blood obtained from a single patient. Results of mRNA expression are expressed as Ct's in this example, but may be expressed as, e.g., relative fluorescence units, copy number or any other quantifiable, precise and calibrated form, for the genes IL1A, IL1B, TNF, IFNG and IL10. From the gene expression values, the acute inflammation values were determined algebraically according in proportion to the expression 1/4 ⁇ IL1A ⁇ +1/4 ⁇ IL1B ⁇ +1/4 ⁇ TNF ⁇ +1/4 ⁇ INFG ⁇ 1/ ⁇ IL10 ⁇ .
  • FIGS. 6 and 7 show the arithmetic mean values for gene expression profiles (using the 48 loci of the Inflammation Gene Expression Panel (Precision ProfileTM for Inflammatory Response)) obtained from whole blood of two distinct patient populations (patient sets). These patient sets are both normal or undiagnosed.
  • the first patient set which is identified as Bonfils (the plot points for which are represented by diamonds), is composed of 17 subjects accepted as blood donors at the Bonfils Blood Center in Denver, Colo.
  • the second patient set is 9 donors, for which Gene Expression Profiles were obtained from assays conducted four times over a four-week period. Subjects in this second patient set (plot points for which are represented by squares) were recruited from employees of Source Precision Medicine, Inc., the assignee herein.
  • Gene expression averages for each population were calculated for each of 48 gene loci of the Gene Expression Inflammation Panel.
  • the results for loci 1-24 (sometimes referred to below as the Inflammation 48A loci) are shown in FIG. 6 and for loci 25-48 (sometimes referred to below as the Inflammation 48B loci) are shown in FIG. 7 .
  • FIG. 8 shows arithmetic mean values for gene expression profiles
  • patient sets also obtained from whole blood of two distinct patient populations (patient sets).
  • One patient set, expression values for which are represented by triangular data points, is 24 normal, undiagnosed subjects (who therefore have no known inflammatory disease).
  • the other patient set, the expression values for which are represented by diamond-shaped data points, is four patients with rheumatoid arthritis and who have failed therapy (who therefore have unstable rheumatoid arthritis).
  • FIG. 9 shows the shows arithmetic mean values for gene expression profiles using 24 loci of the Inflammation Gene Expression Panel (Precision ProfileTM for Inflammatory Response)) also obtained from whole blood of two distinct patient sets.
  • One patient set, expression values for which are represented by diamond-shaped data points, is 17 normal, undiagnosed subjects (who therefore have no known inflammatory disease) who are blood donors.
  • the other patient set, the expression values for which are represented by square-shaped data points is 16 subjects, also normal and undiagnosed, who have been monitored over six months, and the averages of these expression values are represented by the square-shaped data points.
  • the cross-sectional gene expression-value averages of a first healthy population match closely the longitudinal gene expression-value averages of a second healthy population, with approximately 7% or less variation in measured expression value on a gene-to-gene basis.
  • FIG. 10 shows the shows gene expression values (using 14 loci of the Inflammation Gene Expression Panel (Precision ProfileTM for Inflammatory Response)) obtained from whole blood of 44 normal undiagnosed blood donors (data for 10 subjects of which is shown). Again, the gene expression values for each member of the population (set) are closely matched to those for the entire set, represented visually by the consistent peak heights for each of the gene loci. Other subjects of the set and other gene loci than those depicted here display results that are consistent with those shown here.
  • population normative values for a Gene Expression Profile can be used in comparative assessment of individual subjects as to biological condition, including both for purposes of health and/or disease.
  • the normative values for a Gene Expression Profile may be used as a baseline in computing a “calibrated profile data set” (as defined at the beginning of this section) for a subject that reveals the deviation of such subject's gene expression from population normative values.
  • Population normative values for a Gene Expression Profile can also be used as baseline values in constructing index functions in accordance with embodiments of the present invention. As a result, for example, an index function can be constructed to reveal not only the extent of an individual's inflammation expression generally but also in relation to normative values.
  • FIG. 11 shows the expression levels for each of four genes (of the Inflammation Gene Expression Panel (Precision ProfileTM for Inflammatory Response)), of a single subject, assayed monthly over a period of eight months. It can be seen that the expression levels are remarkably consistent over time.
  • FIGS. 12 and 13 similarly show in each case the expression levels for each of 48 genes (of the Inflammation Gene Expression Panel), of distinct single subjects (selected in each case on the basis of feeling well and not taking drugs), assayed, in the case of FIG. 12 weekly over a period of four weeks, and in the case of FIG. 13 monthly over a period of six months.
  • the expression levels are remarkably consistent over time, and also similar across individuals.
  • FIG. 14 also shows the effect over time, on inflammatory gene expression in a single human subject, of the administration of an anti-inflammatory steroid, as assayed using the Inflammation Gene Expression Panel (Precision ProfileTM for Inflammatory Response).
  • 24 of 48 loci are displayed.
  • the subject had a baseline blood sample drawn in a PAX RNA isolation tube and then took a single 60 mg dose of prednisone, an anti-inflammatory, prescription steroid. Additional blood samples were drawn at 2 hr and 24 hr post the single oral dose. Results for gene expression are displayed for all three time points, wherein values for the baseline sample are shown as unity on the x-axis.
  • FIG. 15 in a manner analogous to FIG. 14 , shows the effect over time, via whole blood samples obtained from a human subject, administered a single dose of prednisone, on expression of 5 genes (of the Inflammation Gene Expression Panel (Precision ProfileTM for Inflammatory Response)).
  • Each whole blood sample was challenged by the addition of 0.1 ng/ml of lipopolysaccharide (a Gram-negative endotoxin) and a gene expression profile of the sample, post-challenge, was determined.
  • FIG. 16 also shows the effect over time, on inflammatory gene expression in a single human subject suffering from rheumatoid arthritis, of the administration of a TNF-inhibiting compound, but here the expression is shown in comparison to the cognate locus average previously determined (in connection with FIGS. 6 and 7 ) for the normal (i.e., undiagnosed, healthy) patient set.
  • the subject was followed over a twelve-week period. The subject was enrolled in the study because of a failure to respond to conservative drug therapy for rheumatoid arthritis and a plan to change therapy and begin immediate treatment with a TNF-inhibiting compound.
  • Blood was drawn from the subject prior to initiation of new therapy (visit 1). After initiation of new therapy, blood was drawn at 4 weeks post change in therapy (visit 2), 8 weeks (visit 3), and 12 weeks (visit 4) following the start of new therapy. Blood was collected in PAX RNA isolation tubes, held at room temperature for two hours and then frozen at ⁇ 30° C.
  • Frozen samples were shipped to the central laboratory at Source Precision Medicine, the assignee herein, in Boulder, Colo. for determination of expression levels of genes in the 48-gene Inflammation Gene Expression Panel (Precision ProfileTM for Inflammatory Response).
  • the blood samples were thawed and RNA extracted according to the manufacturer's recommended procedure. RNA was converted to cDNA and the level of expression of the 48 inflammatory genes was determined. Expression results are shown for 11 of the 48 loci in FIG. 16 . When the expression results for the 111 loci are compared from visit one to a population average of normal blood donors from the United States, the subject shows considerable difference. Similarly, gene expression levels at each of the subsequent physician visits for each locus are compared to the same normal average value.
  • FIG. 17A further illustrates the consistency of inflammatory gene expression, illustrated here with respect to 7 loci of (of the Inflammation Gene Expression Panel (Precision ProfileTM for Inflammatory Response)), in a set of 44 normal, undiagnosed blood donors. For each individual locus is shown the range of values lying within ⁇ 2 standard deviations of the mean expression value, which corresponds to 95% of a normally distributed population. Notwithstanding the great width of the confidence interval (95%), the measured gene expression value ( ⁇ CT)—remarkably—still lies within 10% of the mean, regardless of the expression level involved. As described in further detail below, for a given biological condition an index can be constructed to provide a measurement of the condition.
  • ⁇ CT measured gene expression value
  • an inflammation index value was determined for each member of a set of 42 normal undiagnosed blood donors, and the resulting distribution of index values, shown in the figure, can be seen to approximate closely a normal distribution, notwithstanding the relatively small subject set size.
  • the values of the index are shown relative to a O-based median, with deviations from the median calibrated in standard deviation units. Thus 90% of the subject set lies within +1 and ⁇ 1 of a 0 value.
  • FIG. 17C illustrates the use of the same index as FIG. 17B , where the inflammation median for a normal population of subjects has been set to zero and both normal and diseased subjects are plotted in standard deviation units relative to that median.
  • An inflammation index value was determined for each member of a normal, undiagnosed population of 70 individuals (black bars). The resulting distribution of index values, shown in FIG. 17C , can be seen to approximate closely a normal distribution.
  • index values were calculated for individuals from two diseased population groups, (1) rheumatoid arthritis patients treated with methotrexate (MTX) who are about to change therapy to more efficacious drugs (e.g., TNF inhibitors) (hatched bars), and (2) rheumatoid arthritis patients treated with disease modifying anti-rheumatoid drugs (DMARDS) other than MTX, who are about to change therapy to more efficacious drugs (e.g., MTX).
  • MTX methotrexate
  • DARDS disease modifying anti-rheumatoid drugs
  • FIG. 18 plots, in a fashion similar to that of FIG. 17A , Gene Expression Profiles, for the same 7 loci as in FIG. 17A , two different 6-subject populations of rheumatoid arthritis patients.
  • One population (called “stable” in the figure) is of patients who have responded well to treatment and the other population (called “unstable” in the figure) is of patients who have not responded well to treatment and whose therapy is scheduled for change. It can be seen that the expression values for the stable patient population, lie within the range of the 95% confidence interval, whereas the expression values for the unstable patient population for 5 of the 7 loci are outside and above this range.
  • the right-hand portion of the figure shows an average inflammation index of 9.3 for the unstable population and an average inflammation index of 1.8 for the stable population, compared to 1 for a normal undiagnosed population of patients.
  • the index thus provides a measure of the extent of the underlying inflammatory condition, in this case, rheumatoid arthritis.
  • the index besides providing a measure of biological condition, can be used to measure the effectiveness of therapy as well as to provide a target for therapeutic intervention.
  • FIG. 19 thus illustrates use of the inflammation index for assessment of a single subject suffering from rheumatoid arthritis, who has not responded well to traditional therapy with methotrexate.
  • the inflammation index for this subject is shown on the far right at start of a new therapy (a TNF inhibitor), and then, moving leftward, successively, 2 weeks, 6 weeks, and 12 weeks thereafter.
  • the index can be seen moving towards normal, consistent with physician observation of the patient as responding to the new treatment.
  • FIG. 20 similarly illustrates use of the inflammation index for assessment of three subjects suffering from rheumatoid arthritis, who have not responded well to traditional therapy with methotrexate, at the beginning of new treatment (also with a TNF inhibitor), and 2 weeks and 6 weeks thereafter.
  • the index in each case can again be seen moving generally towards normal, consistent with physician observation of the patients as responding to the new treatment.
  • FIGS. 21-23 shows the inflammation index for an international group of subjects, suffering from rheumatoid arthritis, each of whom has been characterized as stable (that is, not anticipated to be subjected to a change in therapy) by the subjects' treating physician.
  • FIG. 21 shows the index for each of 10 patients in the group being treated with methotrexate, which known to alleviate symptoms without addressing the underlying disease.
  • FIG. 22 shows the index for each of 10 patients in the group being treated with Enbrel (an TNF inhibitor), and
  • FIG. 23 shows the index for each 10 patients being treated with Remicade (another TNF inhibitor). It can be seen that the inflammation index for each of the patients in FIG. 21 is elevated compared to normal, whereas in FIG.
  • the patients being treated with Enbrel as a class have an inflammation index that comes much closer to normal (80% in the normal range).
  • FIG. 23 it can be seen that, while all but one of the patients being treated with Remicade have an inflammation index at or below normal, two of the patients have an abnormally low inflammation index, suggesting an immunosuppressive response to this drug. (Indeed, studies have shown that Remicade has been associated with serious infections in some subjects, and here the immunosuppressive effect is quantified.)
  • one subject has an inflammation index that is significantly above the normal range. This subject in fact was also on a regimen of an anti-inflammation steroid (prednisone) that was being tapered; within approximately one week after the inflammation index was sampled, the subject experienced a significant flare of clinical symptoms.
  • prednisone anti-inflammation steroid
  • these examples show a measurement, derived from the assay of blood taken from a subject, pertinent to the subject's arthritic condition. Given that the measurement pertains to the extent of inflammation, it can be expected that other inflammation-based conditions, including, for example, cardiovascular disease, may be monitored in a similar fashion.
  • FIG. 24 illustrates use of the inflammation index for assessment of a single subject suffering from inflammatory bowel disease, for whom treatment with Remicade was initiated in three doses.
  • the graphs show the inflammation index just prior to first treatment, and then 24 hours after the first treatment; the index has returned to the normal range.
  • the index was elevated just prior to the second dose, but in the normal range prior to the third dose.
  • the index besides providing a measure of biological condition, is here used to measure the effectiveness of therapy (Remicade), as well as to provide a target for therapeutic intervention in terms of both dose and schedule.
  • FIG. 25 shows Gene Expression Profiles with respect to 24 loci (of the Inflammation Gene Expression Panel (Precision ProfileTM for Inflammatory Response)) for whole blood treated with Ibuprofen in vitro in relation to other non-steroidal anti-inflammatory drugs (NSAIDs).
  • the profile for Ibuprofen is in front. It can be seen that all of the NSAIDs, including Ibuprofen share a substantially similar profile, in that the patterns of gene expression across the loci are similar. Notwithstanding these similarities, each individual drug has its own distinctive signature.
  • FIG. 26 illustrates how the effects of two competing anti-inflammatory compounds can be compared objectively, quantitatively, precisely, and reproducibly.
  • expression of each of a panel of two genes (of the Inflammation Gene Expression Panel (Precision ProfileTM for Inflammatory Response)) is measured for varying doses (0.08-250 ⁇ g/ml) of each drug in vitro in whole blood.
  • the market leader drug shows a complex relationship between dose and inflammatory gene response. Paradoxically, as the dose is increased, gene expression for both loci initially drops and then increases in the case the case of the market leader. For the other compound, a more consistent response results, so that as the dose is increased, the gene expression for both loci decreases more consistently.
  • FIGS. 27 through 41 illustrate the use of gene expression panels in early identification and monitoring of infectious disease. These figures plot the response, in expression products of the genes indicated, in whole blood, to the administration of various infectious agents or products associated with infectious agents. In each figure, the gene expression levels are “calibrated”, as that term is defined herein, in relation to baseline expression levels determined with respect to the whole blood prior to administration of the relevant infectious agent. In this respect the figures are similar in nature to various figures of our below-referenced patent application WO 01/25473 (for example, FIG. 15 therein).
  • the concentration change is shown ratiometrically, and the baseline level of 1 for a particular gene locus corresponds to an expression level for such locus that is the same, monitored at the relevant time after addition of the infectious agent or other stimulus, as the expression level before addition of the stimulus.
  • Ratiometric changes in concentration are plotted on a logarithmic scale. Bars below the unity line represent decreases in concentration and bars above the unity line represent increases in concentration, the magnitude of each bar indicating the magnitude of the ratio of the change.
  • FIG. 27 uses a novel bacterial Gene Expression Panel of 24 genes, developed to discriminate various bacterial conditions in a host biological system.
  • Two different stimuli are employed: lipotechoic acid (LTA), a gram positive cell wall constituent, and lipopolysaccharide (LPS), a gram negative cell wall constituent.
  • LTA lipotechoic acid
  • LPS lipopolysaccharide
  • the final concentration immediately after administration of the stimulus was 100 ng/mL, and the ratiometric changes in expression, in relation to pre-administration levels, were monitored for each stimulus 2 and 6 hours after administration. It can be seen that differential expression can be observed as early as two hours after administration, for example, in the IFN ⁇ 2 locus, as well as others, permitting discrimination in response between gram positive and gram negative bacteria.
  • FIG. 28 shows differential expression for a single locus, IFNG, to LTA derived from three distinct sources: S. pyrogenes, B. subtilis , and S. aureus .
  • Each stimulus was administered to achieve a concentration of 100 ng/mL, and the response was monitored at 1, 2, 4, 6, and 24 hours after administration.
  • the results suggest that Gene Expression Profiles can be used to distinguish among different infectious agents, here different species of gram positive bacteria.
  • FIGS. 29 and 30 show the response of the Inflammation 48A and 48B loci respectively (discussed above in connection with FIGS. 6 and 7 respectively) in whole blood to administration of a stimulus of S. aureus and of a stimulus of E. coli (in the indicated concentrations, just after administration, of 10 7 and 10 6 CFU/mL respectively), monitored 2 hours after administration in relation to the pre-administration baseline.
  • the figures show that many of the loci respond to the presence of the bacterial infection within two hours after infection.
  • FIGS. 31 and 32 correspond to FIGS. 29 and 30 respectively and are similar to them, with the exception that the monitoring here occurs 6 hours after administration. More of the loci are responsive to the presence of infection. Various loci, such as IL2, show expression levels that discriminate between the two infectious agents.
  • FIG. 33 shows the response of the Inflammation 48A loci to the administration of a stimulus of E. Coli (again in the concentration just after administration of 10 CFU/mL) and to the administration of a stimulus of an E. coli filtrate containing E. coli bacteria by products but lacking E. coli bacteria.
  • the responses were monitored at 2, 6, and 24 hours after administration. It can be seen, for example, that the responses over time of loci IL1B, IL18 and CSF3 to E. coli and to E. coli filtrate are different.
  • FIG. 34 is similar to FIG. 33 , but here the compared responses are to stimuli from E. coli filtrate alone and from E. coli filtrate to which has been added polymyxin B, an antibiotic known to bind to lipopolysaccharide (LPS).
  • polymyxin B an antibiotic known to bind to lipopolysaccharide (LPS).
  • LPS lipopolysaccharide
  • FIG. 35 illustrates the responses of the Inflammation 48A loci over time of whole blood to a stimulus of S. aureus (with a concentration just after administration of 10 CFU/mL) monitored at 2, 6, and 24 hours after administration. It can be seen that response over time can involve both direction and magnitude of change in expression. (See for example, IL5 and IL18.)
  • FIGS. 36 and 37 show the responses, of the Inflammation 48A and 48B loci respectively, monitored at 6 hours to stimuli from E. coli (at concentrations of 10 6 and 10 2 CFU/mL immediately after administration) and from S. aureus (at concentrations of 10 7 and 10 2 CFU/mL immediately after administration). It can be seen, among other things, that in various loci, such as B7 ( FIG. 36 ), TACI, PLA2G7, and C1QA ( FIG. 37 ), E. coli produces a much more pronounced response than S. aureus . The data suggest strongly that Gene Expression Profiles can be used to identify with high sensitivity the presence of gram negative bacteria and to discriminate against gram positive bacteria.
  • FIGS. 38 and 39 show the responses, of the Inflammation 48B and 48A loci respectively, monitored 2, 6, and 24 hours after administration, to stimuli of high concentrations of S. aureus and E. coli respectively (at respective concentrations of 10 7 and 10 6 CFU/mL immediately after administration).
  • the responses over time at many loci involve changes in magnitude and direction.
  • FIG. 40 is similar to FIG. 39 , but shows the responses of the Inflammation 48B loci.
  • FIG. 41 similarly shows the responses of the Inflammation 48A loci monitored at 24 hours after administration to stimuli high concentrations of S. aureus and E. coli respectively (at respective concentrations of 10 7 and 10 6 CFU/mL immediately after administration).
  • responses at some loci such as GRO1 and GRO2, discriminate between type of infection.
  • FIG. 42 illustrates application of a statistical T-test to identify potential members of a signature gene expression panel that is capable of distinguishing between normal subjects and subjects suffering from unstable rheumatoid arthritis.
  • the grayed boxes show genes that are individually highly effective (t test P values noted in the box to the right in each case) in distinguishing between the two sets of subjects, and thus indicative of potential members of a signature gene expression panel for rheumatoid arthritis.
  • FIG. 43 illustrates, for a panel of 17 genes, the expression levels for 8 patients presumed to have bacteremia. The data are suggestive of the prospect that patients with bacteremia have a characteristic pattern of gene expression.
  • FIG. 44 illustrates application of a statistical T-test to identify potential members of a signature gene expression panel that is capable of distinguishing between normal subjects and subjects suffering from bacteremia.
  • the grayed boxes show genes that are individually highly effective (t test P values noted in the box to the right in each case) in distinguishing between the two sets of subjects, and thus indicative of potential members of a signature gene expression panel for bacteremia.
  • FIG. 45 illustrates application of an algorithm (shown in the figure), providing an index pertinent to rheumatoid arthritis (RA) as applied respectively to normal subjects, RA patients, and bacteremia patients.
  • the index easily distinguishes R A subjects from both normal subjects and bacteremia subjects.
  • FIG. 46 illustrates application of an algorithm (shown in the figure), providing an index pertinent to bacteremia as applied respectively to normal subjects, rheumatoid arthritis patients, and bacteremia patients.
  • the index easily distinguishes bacteremia subjects from both normal subjects and rheumatoid arthritis subjects.
  • Results of high precision gene expression analysis are shown below in FIG. 47 .
  • the “y” axis reports the gene expression level in standard deviation units compared to the Source Precision Medicine Normal Reference Population Value for that gene locus at dates May 22, 2002 (before prednisone treatment), May 28, 2002 (after 5 mg treatment on May 22) and Jul. 15, 2002 (after 100 mg prednisone treatment on May 28, tapering to 5 mg within one week).
  • Expression Results for several genes from the 72 gene locus Multiple Sclerosis Precision Profile are shown along the “x” axis.
  • Some gene loci for example IL18; IL1B; MMP9; PTGS2, reflect the severity of the signs while other loci, for example IL10, show effects induced by the steroid treatment. Other loci reflect the non-relapsing TIMP1; TNF; HMOX1.
  • RRMS relapsing remitting multiple sclerosis
  • Selected markers were tested in additional trials in patients known to have MS, and those suspected of having MS.
  • genes selected to be especially probative in characterizing MS and inflammation related to MS such conditions are identified in patients using the herein-described gene expression profile techniques and methods of characterizing multiple sclerosis or inflammatory conditions related to multiple sclerosis in a subject based on a sample from the subject.
  • RRMS subjects experiencing a clinical exacerbation showed altered inflammatory-immune response gene expression compared to RRMS patients during remission and healthy subjects. Additionally, gene expression changes are evident in patients who have exacerbations coincident with initiation and completion of treatment.
  • This system thus provides a gene expression assay system for monitoring MS patients that is predictive of disease progression and treatment responsiveness.
  • gene expression profile data sets were determined and prepared from inflammation and immune-response related genes (mRNA and protein) in whole blood samples taken from RRMS patients before, during and after clinical exacerbation. Samples taken during an exacerbation were collected prior to treatment for the attack. Gene expression results were then correlated with relevant clinical indices as described.
  • the observed data in the gene expression profile data sets was compared to reference profile data sets determined from samples from undiagnosed healthy subjects (normals), gene expression profiles for other chronic immune-related genes, and to profile data sets determined for the individual patients during and after the attack. If desired, a subset of the selected identified genes is coupled with appropriate predictive biomedical algorithms for use in predicting and monitoring RRMS disease activity.
  • a clinical exacerbation is defined as the appearance of a new symptom or worsening/reoccurrence of an old symptom, attributed to RRMS, lasting at least 24 hours in the absence of fever, and preceded by stability or improvement for at least 30 days.
  • Each subject was asked to provide a complete medical history including any existing laboratory test results (i.e. MRI, EDSS scores, blood chemistry, hematology, etc) relevant to the patient's MS contained within the patient's medical records. Additional test results (ordered while the subject is enrolled in the study) relating to the treatment of the patient's MS were collected and correlated with gene expression analysis.
  • any existing laboratory test results i.e. MRI, EDSS scores, blood chemistry, hematology, etc
  • Blood samples for gene expression analysis were collected at screening/baseline (prior to initiation of drug), several times during the treatment phase and several times during follow-up (post-treatment phase). Gene expression results were compared within subjects, between subjects, and to Source Precision Medicine profile data sets determined to be what are termed “Normals”—i.e., a baseline profile dataset determined for a population of healthy (undiagnosed) individuals who do not have MS or other inflammatory conditions, disease, infections. The results were also evaluated to compare and contrast gene expression between different timepoints. This study was used to track individual and population response to the drug, and to correlate clinical symptoms (i.e. disease progression, disease remittance, adverse events) with gene expression.
  • Normal i.e., a baseline profile dataset determined for a population of healthy (undiagnosed) individuals who do not have MS or other inflammatory conditions, disease, infections.
  • the results were also evaluated to compare and contrast gene expression between different timepoints. This study was used to track individual and population response to the drug, and to correlate clinical symptoms (i.
  • Baseline samples from a subset of patients were analyzed.
  • the preliminary data from the baseline samples suggest that that only a plurality of or optionally several specific genetic markers is required to identify MS across a population of samples.
  • the study was also used to track drug response and clinical endpoints.
  • the gene expression profile data sets were then assessed for their ability to track individual response to therapy, for identifying a subset of genes that exhibit altered gene expression in MS and/or are affected by the drug treatment.
  • Clinical data collected during the study include: MRIs, disease progression tests (EDSS, MSFC, ambulation tests, auditory testing, dexterity testing), medical history, concomitant medications, adverse events, physical exam, hematology and chemistry labs, urinalysis, and immunologic testing.
  • Subjects enrolled in the study were asked to discontinue any MS disease modifying therapies they may be using for their disease for at least 3 months prior to dosing with the study drug or drugs.
  • FIGS. 48 through 53 show various analyses of data performed using latent class modeling. From a targeted 104-gene panel, selected to be informative relative to biological state of MS patients (shown in Table 4), primers and probes were prepared for a subset of 54 genes (shown in Table 1B) (those with p-values of 0.05 or better) or 72 genes (shown in Tables 1B and 2 combined). Gene expression profiles were obtained using these subsets of genes, and of these individual genes, ITGAM was found to be uniquely and increasingly informative regarding MS, yielding the best discrimination from normals of the genes examined.
  • Table 1A In order, ranked by increasing p-values, with higher values indicating less discrimination from normals, the following genes shown in Table 1A were determined to be especially useful in discriminating MS subjects (all MS and 3-month washed out MS) from normals (listed below from more discriminating to less discriminating). A ranking of the top 54 genes is shown in Table 1B, listed from more discriminating to less discriminating, by p-value.
  • ITGAM was shown to be most discriminating for MS, have the lowest p-value of all genes examined.
  • Latent Class Modeling was then performed with several other genes in combination with ITGAM, to produce three-gene models, four-gene models, and 5-gene models for characterizing MS relative to normals data for a variety of MS subjects. These results are shown in FIGS. 48 through 53 , discussed below.
  • FIG. 48 shows a three-gene model generated with Latent Class Modeling using ITGAM in combination with MMP9 and ITGA4.
  • four different groups of MS subjects were compared to normals data for a subset of 72 genes of the 104-gene panel shown in Table 4.
  • the question asked was, using only ITGAM combined with two other genes, in this case, MMP9 and ITGA4, is it possible to discriminate MS subjects from normal subjects (those with no history or diagnosis of MS)
  • the groups of MS patients included “washed-out” subjects, i.e. those diagnosed with MS but off any treatment for three months or longer, and who are represented by Xs and diamonds.
  • Another group of subjects were MS subjects who were not washed out from treatment, but rather were on a treatment regimen at the time of this study.
  • the subjects represented by circles were subjects from another clinical study diagnosed with MS and who were also on a treatment regimen at the time of this study.
  • two subjects “flared” during the study and were put on different therapies, and thus moved towards the normal range, as indicated by data taken at that later time and represented in this figures as the star (mf10) and the flower (mf8).
  • Normals data are represented by pentagons.
  • FIG. 49 shows a scatter plot for an alternative three-gene model using ITGAM combined with CD4 and MMP9.
  • the groups of MS patients included “washed out” subjects (Xs), subjects from one clinical study on a treatment regimen (triangle), subjects from another clinical study on a treatment regimen (squares), subjects on an experimental treatment regimen (diamonds), two subjects who flared during the study (mf8 and mf10), and normal subjects (circles).
  • Xs “washed out” subjects
  • subjects from one clinical study on a treatment regimen triangle
  • subjects from another clinical study on a treatment regimen squares
  • subjects on an experimental treatment regimen diamonds
  • two subjects who flared during the study mf8 and mf10
  • normal subjects circles
  • FIG. 50 shows a scatter plot of the same alternative three-gene model of FIG. 49 using ITGAM with MMP9 and CD4 but now displaying only washed out subjects relative to normals. As indicated by the straight line, there is almost complete discrimination with this model between normals (circles) and “washed out” (Xs) subjects.
  • FIG. 51 shows a scatter plot of a four-gene model useful for discriminating all MS subjects, whether washed out, on treatment, or pre-diagnosis.
  • the four-gene model was produced using Latent Class Modeling with ITGAM with ITGA4, MMP9 and CALCA. As can be seen, most MS subjects analyzed (square, diamonds, circles) were quite well-discriminated from normals (pentagon) with this model.
  • FIG. 52 shows a scatter plot of a five-gene model using ITGAM with ITGA4, NFKB1B, MMP9 and CALCA which further discriminates all MS subjects (square diamonds, Xs) from normals (circles). Note that subjects designated as mf10 and mf8 can be seen to move closer to normal upon treatment during the study from their “flared” state which occurred after enrollment.
  • FIG. 53 shows a scatter plot of another five-gene model using ITGAM with ITGA4, NFKB1B, MMP9 and CXCR3 replacing CALCA. Because CALCA is a low expression gene in general, an alternative five-gene model was produced replacing CALCA with CXCR3. Again one can see how the two flared subjects, mf10 and mf8 move closer to normals (star and flower) after treatment. Normals (pentagon).
  • Gene Expression Profiles with sufficient precision and calibration as described herein (1) can determine subsets of individuals with a known biological condition, particularly individuals with multiple sclerosis or individuals with inflammatory conditions related to multiple sclerosis; (2) may be used to monitor the response of patients to therapy; (3) may be used to assess the efficacy and safety of therapy; and (4) may used to guide the medical management of a patient by adjusting therapy to bring one or more relevant Gene Expression Profiles closer to a target set of values, which may be normative values or other desired or achievable values. It has been shown that Gene Expression Profiles may provide meaningful information even when derived from ex vivo treatment of blood or other tissue. It has been shown that Gene Expression Profiles derived from peripheral whole blood are informative of a wide range of conditions neither directly nor typically associated with blood.
  • Gene Expression Profiles are used for characterization and monitoring of treatment efficacy of individuals with multiple sclerosis, or individuals with inflammatory conditions related to multiple sclerosis.
  • Gene Expression Profiles are also used for characterization and early identification (including pre-symptomatic states) of infectious disease. This characterization includes discriminating between infected and uninfected individuals, bacterial and viral infections, specific subtypes of pathogenic agents, stages of the natural history of infection (e.g., early or late), and prognosis. Use of the algorithmic and statistical approaches discussed above to achieve such identification and to discriminate in such fashion is within the scope of various embodiments herein.
  • primers and probes were prepared for a subset of 24 genes identified in the Stepwise Regression Analysis shown in Table 3.
  • the top 8 genes (lowest p-value discriminating between MS and Normals) were subject to further analysis in a two-gene model. Each of the top 8 genes, one at a time, was used as the first gene in a 2-gene model, where all 23 remaining genes were evaluated as the second gene in this 2-gene model. (See Table 6). Column four illustrates the evaluated correct classification rates for these models (Data for those combinations of genes that fell below the 75%/75% cutoff, not all shown).
  • the p-values in the 2-gene models assess the fit of the null hypothesis that the 2-gene model yields predictions of class memberships (MS vs. Normal) that are no different from chance predictions. The p-values were obtained from the SEARCH stepwise logistic procedure in the GOLDMineR program.
  • each of the remaining 22 genes was evaluated as being included as a third gene in the model.
  • Table 7 lists these along with the incremental p-value associated with the 3 rd gene. Only models where the incremental p-value ⁇ 0.05 are listed. The others were excluded because the additional MS vs. Normal discrimination associated with the 3 rd gene was not significant at the 0.05 level.
  • Each of these 3-gene models was evaluated further to determine whether incremental p-values associated with the other 2 genes was also significant. If the incremental p-value of any one of the 3 was found to be less than 0.05, it was excluded because it did not make a significant improvement over one of the 2-gene sub-models.
  • TNF inhibitors including ENBREL, HUMERIA, REMICADE, and other agents that inhibit TNF, have been associated with rare cases of new or exacerbated symptoms of demyelinating disorders including but not limited to multiple sclerosis, and optic neuritis, seizure, neuromyelitis optica, transverse myelitis, acute disseminated encephalomyelitis, HIV encephalitis, adrenoleukodystrophy, adrenomyeloneuropathy, progress multifocal leukoencephalopathy, and central pontine myelinolysis, and CNS manifestations of systemic vasculitis, some case presenting with mental status changes and some associated with permanent disability.
  • demyelinating disorders including but not limited to multiple sclerosis, and optic neuritis, seizure, neuromyelitis optica, transverse myelitis, acute disseminated encephalomyelitis, HIV encephalitis, adrenoleukodystrophy, adrenomye
  • TNF inhibitors have been shown to accelerate the demyelination process causing nerve lesions. For example, cases of transverse myelitis, optic neuritis, multiple sclerosis, and new onset or exacerbation of seizure disorders have been observed in association with ENBREL therapy. The causal relationship to ENBREL therapy remains unclear. While no clinical trials have been performed evaluating ENBREL therapy in patients with multiple sclerosis, other TNF antagonists administered to patients with multiple sclerosis have been associated with increases in disease activity. As such, prescribers should exercise caution in considering the use of ENBREL or other anti-TNF therapeutics in patients with preexisting or recent-onset central nervous system demyelinating disorders.
  • the present invention provides a method for predicting an adverse effect from anti-TNF therapy in a subject.
  • the method comprises obtaining a sample from the subject (e.g., blood, tissue, or cell), the sample providing a source of RNAs, assessing a profile data set of a plurality of members, each member being a quantitative measure of the amount of a distinct RNA constituent in a panel of constituents (e.g., two or more constituents from any of Tables 1-10), selected so that measurement of the constituents enables characterization of the presumptive signs of a multiple sclerosis, wherein such measure for each constituent is obtained under measurement conditions that are substantially repeatable to produce a patient data set.
  • This patient data set is then compared to a baseline profile data set, e.g.
  • a patient data set that is similar to the baseline profile data set indicates the subject is at risk for suffering an adverse effect from anti-TNF therapy.
  • the sample is obtained, prior to, during, or after administration of an anti-TNF therapeutic regimen.
  • the method is useful for screening subjects suffering from an inflammatory condition for a demyelinating disease prior to the administration of anti-TNF therapy for the treatment of the inflammatory condition.
  • the inflammatory condition may include but are not limited to rheumatoid arthritis, psoriasis, ankylosing spondylitis, psoriatic arthritis and Crohn's disease.
  • the demyelinating condition may include but is not limited to multiple sclerosis, optic neuritis, seizure, neuromyelitis optica, transverse myelitis, acute disseminated encephalomyelitis, HIV encephalitis, adrenoleukodystrophy, adrenomyeloneuropathy, progress multifocal leukoencephalopathy, and central pontine myelinolysis, and CNS manifestations of systemic vasculitis,
  • Enbrel containing etanercept, is a breakthrough product approved for the treatment of chronic inflammatory diseases such as rheumatoid arthritis, juvenile rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, and psoriasis. Enbrel continues to maintain a leading position in the dermatology and rheumatology biologic marketplaces, ranking No. 1 in worldwide sales among biotechnology products used in rheumatology and dermatology.
  • Abbott's Humira has been approved for treatment of rheumatoid arthritis in 57 countries, and for psoriatic arthritis and early RA in some European countries and the US.
  • Remicade has now achieved approvals in the treatment of such inflammatory diseases as Crohn's disease, rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis.
  • inflammatory diseases as Crohn's disease, rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis.
  • Remicade has been used to treat more than half a million patients worldwide.
  • Rheumatoid arthritis has a worldwide distribution with an estimated prevalence of 1 to 2%. Prevalence increases with age, approaching 5% in women over age 55. US prevalence of 2 million patients is 0.68%. The average annual incidence in the United States is about 70 per 100,000 annually. Over 200,000 new cases in US per year. Both incidence and prevalence of rheumatoid arthritis are two to three times greater in women than in men.
  • Chronic plaque psoriasis represents approximately 80% of people with psoriasis with a US prevalence of approximately 5.7 million (2%). 10-20% of patients with plaque psoriasis also experience psoriatic arthritis.
  • CCL3 Chemokine C—C Cytokines- AKA: MIP1-alpha; monokine that binds to motif
  • ligand 3 chemokines- CCR1, CCR4 and CCR5 major HIV- growth factors suppressive factor produced by CD8 cells.
  • CCL4 Chemokine C—C Cytokines- Inflammatory and chemotactic monokine; Motif) ligand 4 chemokines- binds to CCR5 and CCR8 growth factors
  • CCL5 Chemokine C—C Cytokines- Binds to CCR1, CCR3, and CCR5 and is a Motif)
  • CCR1 chemokine C—C chemokine A member of the beta chemokine receptor motif) receptor 1 receptor family (seven transmembrane protein).
  • CCR3 Chemokine C—C Chemokine C—C type chemokine receptor (Eotaxin motif) receptor 3 receptor receptor) binds to Eotaxin, Eotaxin-3, MCP-3, MCP-4, SCYA5/RANTES and mip-1 delta thereby mediating intracellular calcium flux.
  • Eotaxin motif chemokine receptor 3 receptor receptor
  • CCR5 chemokine (C—C chemokine Binds to CCL3/MIP-1a and CCL5/RANTES. motif) receptor 5 receptor
  • CD14 CD14 antigen Cell Marker LPS receptor used as marker for monocytes CD19 CD19 antigen Cell Marker AKA Leu 12; B cell growth factor CD3Z CD3 antigen, zeta Cell Marker T-cell surface glycoprotein polypeptide CD4 CD4 antigen (p55) Cell Marker Helper T-cell marker CD86 CD 86 Antigen (cD Cell signaling AKA B7-2; membrane protein found in B 28 antigen ligand) and activation lymphocytes and monocytes; co-stimulatory signal necessary for T lymphocyte proliferation through IL2 production.
  • CD14 CD14 antigen Cell Marker LPS receptor used as marker for monocytes CD19 CD19 antigen Cell Marker AKA Leu 12; B cell growth factor CD3Z CD3 antigen,
  • CXCL10 Chemokine C—X—C Cytokines- AKA: Gamma IP10; interferon inducible motif
  • ligand 10 chemokines- cytokine IP10; SCYB10; Ligand for CXCR3; growth factors binding causes stimulation of monocytes, NK cells; induces T cell migration
  • CXCR3 chemokine C—X—C cytokines- Binds to SCYB10/IP-10, SCYB9/MIG, motif
  • receptor 3 chemokines- SCYB11/I-TAC Binding of chemokines to growth factors CXCR3 results in integrin activation, cytoskeletal changes and chemotactic migration.
  • DPP4 Dipeptidyl-peptidase 4 Membrane Removes dipeptides from unmodified, n- protein; terminus prolines; has role in T cell activation exopeptidase DTR Diphtheria toxin cell signaling, Thought to be involved in macrophage- receptor (heparin- mitogen mediated cellular proliferation. DTR is a binding epidermal potent mitogen and chemotactic factor for growth factor-like fibroblasts and smooth muscle cells, but not growth factor) endothelial cells.
  • HLA-DRA Major Membrane Anchored heterodimeric molecule; cell- Histocompatability protein surface antigen presenting complex Complex; class II, DR alpha HMOX1 Heme oxygenase Enzyme/ Endotoxin inducible (decycling) 1 Redox HSPA1A Heat shock protein 70 Cell Signaling heat shock protein 70 kDa; Molecular and activation chaperone, stabilizes AU rich mRNA HIST1H1C Histo 1, Hic Basic nuclear responsible for the nucleosome structure protein within the chromosomal fiber in eukaryotes; may attribute to modification of nitrotyrosine- containing proteins and their immunoreactivity to antibodies against nitrotyrosine.
  • IL1A Interleukin 1, alpha Cytokines- Proinflammatory; constitutively and inducibly chemokines- expressed in variety of cells.
  • growth factors cytosolic and released only during severe inflammatory disease IL1B Interleukin 1, beta Cytokines- Proinflammatory; constitutively and inducibly chemokines- expressed by many cell types, secreted growth factors IL1R1 Interleukin 1 receptor, Cell signaling AKA: CD12 or IL1R1RA; Binds all three type I and activation forms of interleukin-1 (IL1A, IL1B and IL1RA).
  • IRF7 Interferon regulatory Transcription Regulates transcription of interferon genes factor 7 Factor through DNA sequence-specific binding. Diverse roles include virus-mediated activation of interferon, and modulation of cell growth, differentiation, apoptosis, and immune system activity. ITGA-4 integrin alpha 4 integrin receptor for fibronectin and VCAM1; triggers homotypic aggregation for VLA4 positive leukocytes; participates in cytolytic T-cell interactions with target cells.
  • MX1 Myxovirus resistance peptide Cytoplasmic protein induced by influenza; 1; interferon inducible associated with MS protein p78 N33 Putative prostate Tumor Integral membrane protein. Associated with cancer tumor Suppressor homozygous deletion in metastatic prostate suppressor cancer.
  • NFKB1 Nuclear factor of Transcription p105 is the precursor of the p50 subunit of the kappa light Factor nuclear factor NFKB, which binds to the polypeptide gene kappa-b consensus sequence located in the enhancer in B-cells 1 enhancer region of genes involved in immune (p105) response and acute phase reactions; the precursor does not bind DNA itself NFKBIB Nuclear factor of Transcription Inhibits/regulates NFKB complex activity by kappa light Regulator trapping NFKB in the cytoplasm. polypeptide gene Phosphorylated serine residues mark the enhancer in B-cells NFKBIB protein for destruction thereby inhibitor, beta allowing activation of the NFKB complex.
  • PAFAH1B1 Platelet activating Enzyme Inactivates platelet activating factor by factor removing the acetyl group acetylhydrolase, isoform !b, alpha subunit; 45 kDa PF4 Platelet Factor 4 Chemokine PF4 is released during platelet aggregation (SCYB4) and is chemotactic for neutrophils and monocytes.
  • PI3 Proteinase inhibitor 3 Proteinase aka SKALP; Proteinase inhibitor found in skin derived inhibitor- epidermis of several inflammatory skin protein binding- diseases; it's expression can be used as a extracellular marker of skin irritancy matrix PLA2G7 Phospholipase A2, Enzyme/ Platelet activating factor group VII (platelet Redox activating factor acetylhydrolase, plasma) PLAU Plasminogen proteinase AKA uPA; cleaves plasminogen to plasmin (a activator, urokinase protease responsible for nonspecific extracellular matrix degradation; UPA stimulates cell migration via a UPA receptor PLAUR plasminogen Membrane key molecule in the regulation of cell-surface activator, urokinase protein
  • Serine or cysteine
  • SFTPD Surfactant, pulmonary extracellular AKA: PSPD; mannose-binding protein; associated protein D lipoprotein suggested role in innate immunity and surfactant metabolism
  • SPP1 secreted cell signaling binds vitronectin; protein ligand of CD44, phosphoprotein 1 and activation cytokine for type 1 responses mediated by (osteopontin) macrophages STAT3 Signal transduction Transcription AKA
  • metalloproteinase 1 Proteinase metalloproteinases such as collagenase Inhibitor TLR2 toll-like receptor 2 cell signaling mediator of peptidoglycan and lipotechoic and activation acid induced signaling TLR4 Toll-like receptor 4 Cell signaling mediator of
  • TH 1 cytokine Mediates host response to bacterial stimulus; Regulates cell growth & differentiation TNFRSF7 Tumor necrosis factor Membrane Receptor for CD27L; may play a role in receptor superfamily, protein; activation of T cells member 7 receptor TNFSF13B Tumor necrosis factor Cytokines- B cell activating factor, TNF family (ligand) superfamily, chemokines- member 13b growth factors TNFRSF13B Tumor necrosis factor Cytokines- B cell activating factor, TNF family receptor superfamily, chemokines- member 13, subunit growth factors beta TNFSF5 Tumor necrosis factor Cytokines- Ligand for CD40; expressed on the surface of (ligand) superfamily, chemokines- T cells.
  • TNFSF6 Tumor necrosis factor Cytokines- AKA FasL; Ligand for FAS antigen; (ligand) superfamily, chemokines- transduces apoptotic signals into cells member 6 growth factors
  • TREM1 Triggering receptor cell signaling Member of the Ig superfamily; receptor expressed on myeloid and activation exclusively expressed on myeloid cells.
  • cells 1 TREM1 mediates activation of neutrophils and monocytes and may have a predominant role in inflammatory responses VEGF vascular endothelial cytokines- VPF; Induces vascular permeability, growth factor chemokines- endothelial cell proliferation, angiogenesis. growth factors Produced by monocytes

Abstract

The present invention provides methods of characterizing multiple sclerosis or inflammatory conditions associated with multiple sclerosis using gene expression profiling.

Description

    RELATED APPLICATIONS
  • This application is a continuation in part of U.S. Ser. No. 11/454,533, filed Jun. 16, 2006 and a continuation in part of U.S. Ser. No. 11/155,930, filed Jun. 16, 2005 and claims the benefit of U.S. Ser. No. 60/734,681, filed Nov. 7, 2005, U.S. Ser. No. 60/758,933, filed Jan. 13, 2006, and U.S. Ser. No. 60/831,005, filed Jul. 13, 2006, each of which are incorporated herein by reference in their entireties.
  • FIELD OF THE INVENTION
  • The present invention relates generally to the identification of biological markers associated with the identification of multiple sclerosis. More specifically, the invention relates to the use of gene expression data in the identification, monitoring and treatment of subjects receiving anti-TNF therapy.
  • BACKGROUND OF THE INVENTION
  • Multiple sclerosis (MS) is an autoimmune disease that affects the central nervous system (CNS). The CNS consists of the brain, spinal cord, and the optic nerves. Surrounding and protecting the nerve fibers of the CNS is a fatty tissue called myelin, which helps nerve fibers conduct electrical impulses. In MS, myelin is lost in multiple areas, leaving scar tissue called sclerosis. These damaged areas are also known as plaques or lesions. Sometimes the nerve fiber itself is damaged or broken. Myelin not only protects nerve fibers, but makes their job possible. When myelin or the nerve fiber is destroyed or damaged, the ability of the nerves to conduct electrical impulses to and from the brain is disrupted, and this produces the various symptoms of MS. People with MS can expect one of four clinical courses of disease, each of which might be mild, moderate, or severe. These include Relapsing-Remitting, Primary-Progressive, Secondary-Progressive, and Progressive-Relapsing
  • Individuals Progressive-Relapsing MS experience clearly defined flare-ups (also called relapses, attacks, or exacerbations). These are episodes of acute worsening of neurologic function. They are followed by partial or complete recovery periods (remissions) free of disease progression.
  • Individuals with Primary-Progressive MS experience a slow but nearly continuous worsening of their disease from the onset, with no distinct relapses or remissions. However, there are variations in rates of progression over time, occasional plateaus, and temporary minor improvements.
  • Individuals with Secondary-Progressive MS experience an initial period of relapsing-remitting disease, followed by a steadily worsening disease course with or without occasional flare-ups, minor recoveries (remissions), or plateaus.
  • Individuals with Progressive-Relapsing MS experience a steadily worsening disease from the onset but also have clear acute relapses (attacks or exacerbations), with or without recovery. In contrast to relapsing-remitting MS, the periods between relapses are characterized by continuing disease progression.
  • Information on any condition of a particular patient and a patient's response to types and dosages of therapeutic or nutritional agents has become an important issue in clinical medicine today not only from the aspect of efficiency of medical practice for the health care industry but for improved outcomes and benefits for the patients. Thus a need exists for better ways to diagnose and monitor the progression of multiple sclerosis.
  • Currently, the characterization of disease condition related to MS (including diagnosis, staging, monitoring disease progression, monitoring treatment effects on disease activity) is imprecise. Imaging that detects what appears to be plaques in CNS tissue is typically insufficient, by itself, to give a definitive diagnosis of MS. Diagnosis of MS is often made only after both detection of plaques and of clinically evident neuropathy. It is clear that diagnosis of MS is usually made well after initiation of the disease process; i.e., only after detection of a sufficient number of plaques and of clinically evident neurological symptoms. Additionally, staging of MS is typically done by subjective measurements of exacerbation of symptoms, as well of other clinical manifestations. There are difficulties in diagnosis and staging because symptoms vary widely among individuals and change frequently within the individual. Thus, there is the need for tests which can aid in the diagnosis, monitor the progression and staging of MS. This is of particular importance in patients who are recommended to receive anti-TNF therapy as it is known that anti-TNF therapy exacerbates the clinical manifestations of multiple sclerosis.
  • SUMMARY OF THE INVENTION
  • The invention is based in part upon the identification of gene expression profiles associated with multiple sclerosis (MS). Theses genes are referred to herein as MS-associated genes. More specifically, the invention is based upon the surprising discovery that detection of as few as two MS-associated genes is capable of identifying individuals with or without MS with at least 75% accuracy.
  • In various aspects the invention provides a method for determining a profile data set for characterizing a subject with multiple sclerosis or an inflammatory condition related to multiple sclerosis based on a sample from the subject, the sample providing a source of RNAs, by using amplification for measuring the amount of RNA in a panel of constituents including at least 2 constituents from any of Tables 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 and arriving at a measure of each constituent. The profile data set contains the measure of each constituent of the panel.
  • Also provided by the invention is a method of characterizing multiple sclerosis or inflammatory condition related to multiple sclerosis in a subject, based on a sample from the subject, the sample providing a source of RNAs, by assessing a profile data set of a plurality of members, each member being a quantitative measure of the amount of a distinct RNA constituent in a panel of constituents selected so that measurement of the constituents enables characterization of the presumptive signs of a multiple sclerosis.
  • In yet another aspect the invention provides a method of characterizing multiple sclerosis or an inflammatory condition related to multiple sclerosis in a subject, based on a sample from the subject, the sample providing a source of RNAs, by determining a quantitative measure of the amount of at least one constituent from any of Tables 1-10. In yet another aspect the invention provides a method for predicting an adverse effect from anti-TNF therapy in a subject, based on a sample from the subject, the sample providing a source of RNAs, said method comprising: a) assessing a profile data set of a plurality of members, each member being a quantitative measure of the amount of a distinct RNA constituent in a panel of constituents selected so that measurement of the constituents enables characterization of the presumptive signs of multiple sclerosis or an inflammatory condition related to multiple sclerosis, wherein such measure for each constituent is obtained under measurement conditions that are substantially repeatable to produce a patient data set; and b) comparing the patient data set to a baseline profile data set, wherein the baseline profile data set is related to said multiple sclerosis or inflammatory condition related to multiple sclerosis; wherein a similarity between the patient data set and the baseline profile data set indicates a risk of an adverse effect from anti-TNF therapy in the subject.
  • In still another embodiment, the present invention provides a method for predicting an increased risk to an adverse effect from anti-TNF therapy in a subject, based on a sample from the subject, the sample providing a source of RNAs, said method comprising: a) determining a quantitative measure of the amount of at least one constituent of Table 4 or 10 as a distinct RNA constituent, wherein such measure is obtained under measurement conditions that are substantially repeatable to produce a patient data set; and b) comparing the patient data set to a baseline profile data set, wherein the baseline profile data set is related to said multiple sclerosis or inflammatory condition related to multiple sclerosis; wherein a similarity between the patient data set and the baseline profile data set indicates a risk of an adverse effect from anti-TNF therapy in the subject. In one embodiment, the method of predicting an adverse effect of anti-TNF therapy is performed on a subject suffering from an inflammatory condition, including but not limited to rheumatoid arthritis, psoriasis, ankylosing spondylitis, psoriatic arthritis and Crohn's diseases. The method is performed prior to, during, or after administering an anti-TNF therapeutic or anti-TNF therapeutic regimen to the subject. In a preferred embodiment, said method is performed prior to administering an anti-TNF therapeutic to the subject. By increased risk it is meant that treatment with anti-TNF therapy is contraindicated.
  • The panel of constituents are selected so as to distinguish from a normal and a MS-diagnosed subject. The MS-diagnosed subject is washed out from therapy for three or more months. Preferably, the panel of constituents are selected so as to distinguish from a normal and a MS-diagnosed subject with at least 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or greater accuracy. By “accuracy” is meant that the method has the ability to distinguish between subjects having multiple sclerosis or an inflammatory condition associated with multiple sclerosis and those that do not. Accuracy is determined for example by comparing the results of the Gene Expression Profiling to standard accepted clinical methods of diagnosing MS, e.g. MRI, sign and symptoms such as blurred vision, fatigue, loss or balance.
  • Alternatively, the panel of constituents is selected as to permit characterizing severity of MS in relation to normal over time so as to track movement toward normal as a result of successful therapy and away from normal in response to symptomatic flare.
  • The panel contains 10, 8, 5, 4, 3 or fewer constituents. Optimally, the panel of constituents includes ITGAM, HLADRA, CASP9, ITGAL or STAT3. Alternatively, the panel includes ITGAM and i) CD4 and MMP9, ii) ITGA4 and MMP9, iii) ITGA4, MMP9 and CALCA, iv) ITGA4, MMP9 and NFKB1B, v) ITGA4, MMP9, CALCA and CXCR3, or vi) ITGA4, MMP9, NFKB1B and CXCR3. The panel includes two or more constituents from any of Tables 1-10. In one preferred embodiment, the panel includes two or more constituents from Table 4 or 10. In another preferred embodiment, the panel includes three constituents in any combination shown on Table 7. In yet another preferred embodiment, the panel includes any 2, 3, 4, or 5 genes in the combination shown in Tables 6, 7, 8 and 9 respectively. For example the panel contains i) HLADRA and one or more or the following: ITGAL, CASP9, NFKB1B, STAT2, NFKB1, ITGAM, ITGAL, CD4, IL1B, HSPA1A, ICAM1, IFI16, or TGFBR2; ii) CASP9 and one or more of the following VEGFB, CD14 or JUN; iii) ITGAL and one or more of the following: P13, ITGAM or TGFBR2; and iv) STAT3 and CD14.
  • Optionally, assessing may further include comparing the profile data set to a baseline profile data set for the panel. The baseline profile data set is related to the multiple sclerosis or an inflammatory condition related to multiple sclerosis to be characterized. The baseline profile data set is derived from one or more other samples from the same subject, taken when the subject is in a biological condition different from that in which the subject was at the time the first sample was taken, with respect to at least one of age, nutritional history, medical condition, clinical indicator, medication, physical activity, body mass, and environmental exposure, and the baseline profile data set may be derived from one or more other samples from one or more different subjects. In addition, the one or more different subjects may have in common with the subject at least one of age group, gender, ethnicity, geographic location, nutritional history, medical condition, clinical indicator, medication, physical activity, body mass, and environmental exposure. A clinical indicator may be used to assess multiple sclerosis or am inflammatory condition related to multiple sclerosis of the one or more different subjects, and may also include interpreting the calibrated profile data set in the context of at least one other clinical indicator, wherein the at least one other clinical indicator such as blood chemistry, urinalysis, X-ray or other radiological or metabolic imaging technique, other chemical assays, and physical findings.
  • The baseline profile data set may be derived from one or more other samples from the same subject taken under circumstances different from those of the first sample, and the circumstances may be selected from the group consisting of (i) the time at which the first sample is taken, (ii) the site from which the first sample is taken, (iii) the biological condition of the subject when the first sample is taken.
  • The subject has one or more presumptive signs of a multiple sclerosis. Presumptive signs of multiple sclerosis includes for example, altered sensory, motor, visual or proprioceptive system with at least one of numbness or weakness in one or more limbs, often occurring on one side of the body at a time or the lower half of the body, partial or complete loss of vision, frequently in one eye at a time and often with pain during eye movement, double vision or blurring of vision, tingling or pain in numb areas of the body, electric-shock sensations that occur with certain head movements, tremor, lack of coordination or unsteady gait, fatigue, dizziness, muscle stiffness or spasticity, slurred speech, paralysis, problems with bladder, bowel or sexual function, and mental changes such as forgetfulness or difficulties with concentration, relative to medical standards. Alternatively, the subject is at risk of developing multiple sclerosis, for example the subject has a family history of multiple sclerosis or another autoimmune disorder such as for example rheumatoid arthritis, Crohn's disease, or lupus. Optionally, subject is a candidate for anti-TNF therapy.
  • By multiple sclerosis or an inflammatory condition related to multiple sclerosis is meant that the condition is an autoimmune condition, an environmental condition, a viral infection, a bacterial infection, a eukaryotic parasitic infection, or a fungal infection.
  • The sample is any sample derived from a subject which contains RNA. For example the sample is blood, a blood fraction, body fluid, and a population of cells or tissue from the subject.
  • Optionally one or more other samples can be taken over an interval of time that is at least one month between the first sample and the one or more other samples, or taken over an interval of time that is at least twelve months between the first sample and the one or more samples, or they may be taken pre-therapy intervention or post-therapy intervention. In such embodiments, the first sample may be derived from blood and the baseline profile data set may be derived from tissue or body fluid of the subject other than blood. Alternatively, the first sample is derived from tissue or body fluid of the subject and the baseline profile data set is derived from blood.
  • All of the forgoing embodiments are carried out wherein the measurement conditions are substantially repeatable, particularly within a degree of repeatability of better than five percent or more particularly within a degree of repeatability of better than three percent, and/or wherein efficiencies of amplification for all constituents are substantially similar, more particularly wherein the efficiency of amplification is within two percent, and still more particularly wherein the efficiency of amplification for all constituents is less than one percent.
  • Additionally the invention includes storing the profile data set in a digital storage medium. Optionally, storing the profile data set includes storing it as a record in a database.
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
  • Other features and advantages of the invention will be apparent from the following detailed description and claims.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The foregoing features of the invention will be more readily understood by reference to the following detailed description, taken with reference to the accompanying drawings, in which:
  • FIG. 1A shows the results of assaying 24 genes from the Source Inflammation Gene Panel (shown in Table 1 of U.S. Pat. No. 6,692,916, which patent is hereby incorporated by reference; such Panel is hereafter referred to as the Inflammation Gene Expression Panel, and is incorporated into the 96 gene expression panel shown in Table 10, referred to as the Precision Profile™ for Inflammatory Response) on eight separate days during the course of optic neuritis in a single male subject.
  • 1B illustrates use of an inflammation index in relation to the data of FIG. 1A, in accordance with an embodiment of the present invention.
  • FIG. 2 is a graphical illustration of the same inflammation index calculated at 9 different, significant clinical milestones.
  • FIG. 3 shows the effects of single dose treatment with 800 mg of ibuprofen in a single donor as characterized by the index.
  • FIG. 4 shows the calculated acute inflammation index displayed graphically for five different conditions.
  • FIG. 5 shows a Viral Response Index for monitoring the progress of an upper respiratory infection (URI).
  • FIGS. 6 and 7 compare two different populations using Gene Expression Profiles (with respect to the 48 loci of the Inflammation Gene Expression Panel (which is incorporated in the Precision Profile™ for Inflammatory Response shown in Table 10).
  • FIG. 8 compares a normal population with a rheumatoid arthritis population derived from a longitudinal study.
  • FIG. 9 compares two normal populations, one longitudinal and the other cross sectional.
  • FIG. 10 shows the shows gene expression values for various individuals of a normal population.
  • FIG. 11 shows the expression levels for each of four genes of the Inflammation Gene Expression Panel (which is incorporated in the Precision Profile™ for Inflammatory Response shown in Table 10), of a single subject, assayed monthly over a period of eight months.
  • FIG. 12 shows the expression levels for each of 48 genes of the Inflammation Gene Expression Panel, (which is incorporated in the Precision Profile™ for Inflammatory Response shown in Table 10) of distinct single subjects (selected in each case on the basis of feeling well and not taking drugs), assayed weekly over a period of four weeks.
  • FIG. 13 shows the expression levels for each of 48 genes (of the Inflammation Gene Expression Panel (which is incorporated in the Precision Profile™ for Inflammatory Response shown in Table 10), of distinct single subjects (selected in each case on the basis of feeling well and not taking drugs), assayed monthly over a period of six months.
  • FIG. 14 shows the effect over time, on inflammatory gene expression in a single human subject, of the administration of an anti-inflammatory steroid, as assayed using the Inflammation Gene Expression Panel (which is incorporated in the Precision Profile™ for Inflammatory Response shown in Table 10).
  • FIG. 15, shows the effect over time, via whole blood samples obtained from a human subject, administered a single dose of prednisone, on expression of 5 genes (of the Inflammation Gene Expression Panel (which is incorporated in the Precision Profile™ for Inflammatory Response shown in Table 10).
  • FIG. 16 shows the effect over time, on inflammatory gene expression in a single human subject suffering from rheumatoid arthritis, of the administration of a TNF-inhibiting compound, but here the expression is shown in comparison to the cognate locus average previously determined (in connection with FIGS. 6 and 7) for the normal (i.e., undiagnosed, healthy) population.
  • FIG. 17A illustrates the consistency of inflammatory gene expression in a population.
  • FIG. 17B shows the normal distribution of index values obtained from an undiagnosed population.
  • FIG. 17C illustrates the use of the same index as FIG. 17B, where the inflammation median for a normal population has been set to zero and both normal and diseased subjects are plotted in standard deviation units relative to that median.
  • FIG. 18 plots, in a fashion similar to that of FIG. 17A, Gene Expression Profiles, for the same 7 loci as in FIG. 17A, two different (responder v. non-responder) 6-subject populations of rheumatoid arthritis patients.
  • FIG. 19 illustrates use of the inflammation index for assessment of a single subject suffering from rheumatoid arthritis, who has not responded well to traditional therapy with methotrexate.
  • FIG. 20 illustrates use of the inflammation index for assessment of three subjects suffering from rheumatoid arthritis, who have not responded well to traditional therapy with methotrexate.
  • FIG. 21 shows the inflammation index for an international group of subjects, suffering from rheumatoid arthritis, undergoing three separate treatment regimens
  • FIG. 22 shows the inflammation index for an international group of subjects, suffering from rheumatoid arthritis, undergoing three separate treatment regimens
  • FIG. 23 shows the inflammation index for an international group of subjects, suffering from rheumatoid arthritis, undergoing three separate treatment regimens.
  • FIG. 24 illustrates use of the inflammation index for assessment of a single subject suffering from inflammatory bowel disease.
  • FIG. 25 shows Gene Expression Profiles with respect to 24 loci (of the Inflammation Gene Expression Panel (which is incorporated in the Precision Profile™ for Inflammatory Response shown in Table 10) for whole blood treated with Ibuprofen in vitro in relation to other non-steroidal anti-inflammatory drugs (NSAIDs).
  • FIG. 26 illustrates how the effects of two competing anti-inflammatory compounds can be compared objectively, quantitatively, precisely, and reproducibly.
  • FIG. 27 uses a novel bacterial Gene Expression Panel of 24 genes, developed to discriminate various bacterial conditions in a host biological system.
  • FIG. 28 shows differential expression for a single locus, IFNG, to LTA derived from three distinct sources: S. pyrogenes, B. subtilis, and S. aureus.
  • FIG. 29 shows the response after two hours of the Inflammation 48A and 48B loci respectively (discussed above in connection with FIGS. 6 and 7 respectively) in whole blood to administration of a Gram-positive and a Gram-negative organism.
  • FIG. 30 shows the response after two hours of the Inflammation 48A and 48B loci respectively (discussed above in connection with FIGS. 6 and 7 respectively) in whole blood to administration of a Gram-positive and a Gram-negative organism.
  • FIG. 31 shows the response after six hours of the Inflammation 48A and 48B loci respectively (discussed above in connection with FIGS. 6 and 7 respectively) in whole blood to administration of a Gram-positive and a Gram-negative organism.
  • FIG. 32 shows the response after six hours of the Inflammation 48A and 48B loci respectively (discussed above in connection with FIGS. 6 and 7 respectively) in whole blood to administration of a Gram-positive and a Gram-negative organism.
  • FIG. 33 compares the gene expression response induced by E. coli and by an organism-free E. coli filtrate.
  • FIG. 34 is similar to FIG. 33, but compared responses are to stimuli from E. coli filtrate alone and from E. coli filtrate to which has been added polymyxin B.
  • FIG. 35 illustrates the gene expression responses induced by S. aureus at 2, 6, and 24 hours after administration.
  • FIG. 36 illustrates the comparison of the gene expression induced by E. coli and S. aureus under various concentrations and times.
  • FIG. 37 illustrates the comparison of the gene expression induced by E. coli and S. aureus under various concentrations and times.
  • FIG. 38 illustrates the comparison of the gene expression induced by E. coli and S. aureus under various concentrations and times.
  • FIG. 39 illustrates the comparison of the gene expression induced by E. coli and S. aureus under various concentrations and times.
  • FIG. 40 illustrates the comparison of the gene expression induced by E. coli and S. aureus under various concentrations and times.
  • FIG. 41 illustrates the comparison of the gene expression induced by E. coli and S. aureus under various concentrations and times.
  • FIG. 42 illustrates application of a statistical T-test to identify potential members of a signature gene expression panel that is capable of distinguishing between normal subjects and subjects suffering from unstable rheumatoid arthritis.
  • FIG. 43 illustrates for a panel of 17 genes, the expression levels for 8 patients presumed to have bacteremia.
  • FIG. 44 illustrates application of a statistical T-test to identify potential members of a signature gene expression panel that is capable of distinguishing between normal subjects and subjects suffering from bacteremia
  • FIG. 45 illustrates application of an algorithm (shown in the figure), providing an index pertinent to rheumatoid arthritis (RA) as applied respectively to normal subjects, RA patients, and bacteremia patients.
  • FIG. 46 illustrates application of an algorithm (shown in the figure), providing an index pertinent to bacteremia as applied respectively to normal subjects, rheumatoid arthritis patients, and bacteremia patients.
  • FIG. 47 illustrates, for a panel of 47 genes selected genes from Table 1, the expression levels for a patient suffering from multiple sclerosis on dates May 22, 2002 (no treatment), May 28, 2002 (after 5 mg prednisone given on May 22), and Jul. 15, 2002 (after 100 mg prednisone given on May 28, tapering to 5 mg within one week).
  • FIG. 48 shows a scatter plot of a three-gene model useful for discriminating MS subjects generated by Latent Class Modeling analysis using ITGAM with MMP9 and ITGA4.
  • FIG. 49 shows a scatter plot of an alternative three-gene model useful for discriminating MS subjects using ITGAM with CD4 and MMP9.
  • FIG. 50 shows a scatter plot of the same alternative three-gene model of FIG. 49 useful for discriminating MS subjects using ITGAM with MMP9 and CD4 but now displaying only washed out subjects relative to normals.
  • FIG. 51 shows a scatter plot of a four-gene model useful for discriminating MS subjects using ITGAM with ITGA4, MMP9 and CALCA.
  • FIG. 52 shows a scatter plot of a five-gene model useful for discriminating MS subjects using ITGAM with ITGA4, NFKB1B, MMP9 and CALCA.
  • FIG. 53 shows another five-gene model useful for discriminating MS subjects using ITGAM with ITGA4, NFKB1B, MMP9 and CXCR3 replacing CALCA.
  • FIG. 54 show a shows a four-gene model useful for discriminating MS subjects using ITGAL, CASP9, HLADRA and TGFBR2.
  • FIG. 55 show a shows a two-gene model useful for discriminating MS subjects using CASP9 and HLADRA.
  • FIG. 56 show a shows a two-gene model useful for discriminating MS subjects using ITGAL and HLADRA.
  • FIG. 57 show a shows a three-gene model useful for discriminating MS subjects using ITGAL, CASP9, and HLADRA.
  • DETAILED DESCRIPTION OF SPECIFIC EMBODIMENTS Definitions
  • The following terms shall have the meanings indicated unless the context otherwise requires:
  • “Algorithm” is a set of rules for describing a biological condition. The rule set may be defined exclusively algebraically but may also include alternative or multiple decision points requiring domain-specific knowledge, expert interpretation or other clinical indicators.
  • An “agent” is a “composition” or a “stimulus”, as those terms are defined herein, or a combination of a composition and a stimulus.
  • “Amplification” in the context of a quantitative RT-PCR assay is a function of the number of DNA replications that are tracked to provide a quantitative determination of its concentration. “Amplification” here refers to a degree of sensitivity and specificity of a quantitative assay technique. Accordingly, amplification provides a measurement of concentrations of constituents that is evaluated under conditions wherein the efficiency of amplification and therefore the degree of sensitivity and reproducibility for measuring all constituents is substantially similar.
  • “Accuracy” is measure of the strength of the relationship between true values and their predictions. Accordingly, accuracy provided a measurement on how close to a true or accepted value a measurement lies
  • A “baseline profile data set” is a set of values associated with constituents of a Gene Expression Panel resulting from evaluation of a biological sample (or population or set of samples) under a desired biological condition that is used for mathematically normative purposes. The desired biological condition may be, for example, the condition of a subject (or population or set of subjects) before exposure to an agent or in the presence of an untreated disease or in the absence of a disease. Alternatively, or in addition, the desired biological condition may be health of a subject or a population or set of subjects. Alternatively, or in addition, the desired biological condition may be that associated with a population or set of subjects selected on the basis of at least one of age group, gender, ethnicity, geographic location, nutritional history, medical condition, clinical indicator, medication, physical activity, body mass, and environmental exposure.
  • A “biological condition” of a subject is the condition of the subject in a pertinent realm that is under observation, and such realm may include any aspect of the subject capable of being monitored for change in condition, such as health, disease including cancer; autoimmune condition; trauma; aging; infection; tissue degeneration; developmental steps; physical fitness; obesity, and mood. As can be seen, a condition in this context may be chronic or acute or simply transient. Moreover, a targeted biological condition may be manifest throughout the organism or population of cells or may be restricted to a specific organ (such as skin, heart, eye or blood), but in either case, the condition may be monitored directly by a sample of the affected population of cells or indirectly by a sample derived elsewhere from the subject. The term “biological condition” includes a “physiological condition”.
  • “Body fluid” of a subject includes blood, urine, spinal fluid, lymph, mucosal secretions, prostatic fluid, semen, haemolymph or any other body fluid known in the art for a subject.
  • “Calibrated profile data set” is a function of a member of a first profile data set and a corresponding member of a baseline profile data set for a given constituent in a panel.
  • A “clinical indicator” is any physiological datum used alone or in conjunction with other data in evaluating the physiological condition of a collection of cells or of an organism. This term includes pre-clinical indicators.
  • A “composition” includes a chemical compound, a nutraceutical, a pharmaceutical, a homeopathic formulation, an allopathic formulation, a naturopathic formulation, a combination of compounds, a toxin, a food, a food supplement, a mineral, and a complex mixture of substances, in any physical state or in a combination of physical states.
  • To “derive” a profile data set from a sample includes determining a set of values associated with constituents of a Gene Expression Panel either (i) by direct measurement of such constituents in a biological sample or (ii) by measurement of such constituents in a second biological sample that has been exposed to the original sample or to matter derived from the original sample.
  • “Distinct RNA or protein constituent” in a panel of constituents is a distinct expressed product of a gene, whether RNA or protein. An “expression” product of a gene includes the gene product whether RNA or protein resulting from translation of the messenger RNA.
  • A “Gene Expression Panel” is an experimentally verified set of constituents, each constituent being a distinct expressed product of a gene, whether RNA or protein, wherein constituents of the set are selected so that their measurement provides a measurement of a targeted biological condition.
  • A “Gene Expression Profile” is a set of values associated with constituents of a Gene Expression Panel resulting from evaluation of a biological sample (or population or set of samples).
  • A “Gene Expression Profile Inflammatory Index” is the value of an index function that provides a mapping from an instance of a Gene Expression Profile into a single-valued measure of inflammatory condition.
  • The “health” of a subject includes mental, emotional, physical, spiritual, allopathic, naturopathic and homeopathic condition of the subject.
  • “Index” is an arithmetically or mathematically derived numerical characteristic developed for aid in simplifying or disclosing or informing the analysis of more complex quantitative information. A disease or population index may be determined by the application of a specific algorithm to a plurality of subjects or samples with a common biological condition.
  • “Inflammation” is used herein in the general medical sense of the word and may be an acute or chronic; simple or suppurative; localized or disseminated; cellular and tissue response, initiated or sustained by any number of chemical, physical or biological agents or combination of agents.
  • “Inflammatory state” is used to indicate the relative biological condition of a subject resulting from inflammation, or characterizing the degree of inflammation
  • A “large number” of data sets based on a common panel of genes is a number of data sets sufficiently large to permit a statistically significant conclusion to be drawn with respect to an instance of a data set based on the same panel.
  • “Multiple sclerosis” (MS) is a debilitating wasting disease. The disease is associated with degeneration of the myelin sheaths surrounding nerve cells which leads to a loss of motor and sensory function.
  • A “normar” subject is a subject who has not been diagnosed with multiple sclerosis, or one who is not suffering from multiple sclerosis.
  • A “normative” condition of a subject to whom a composition is to be administered means the condition of a subject before administration, even if the subject happens to be suffering from a disease.
  • A “panel” of genes is a set of genes including at least two constituents.
  • A “population of cells” refers to any group of cells wherein there is an underlying commonality or relationship between the members in the population of cells, including a group of cells taken from an organism or from a culture of cells or from a biopsy, for example.
  • A “sample” from a subject may include a single cell or multiple cells or fragments of cells or an aliquot of body fluid, taken from the subject, by means including venipuncture, excretion, ejaculation, massage, biopsy, needle aspirate, lavage sample, scraping, surgical incision or intervention or other means known in the art.
  • A “set” or “population” of samples or subjects refers to a defined or selected group of samples or subjects wherein there is an underlying commonality or relationship between the members included in the set or population of samples or subjects.
  • A “Signature Profile” is an experimentally verified subset of a Gene Expression Profile selected to discriminate a biological condition, agent or physiological mechanism of action.
  • A “Signature Panel” is a subset of a Gene Expression Panel, the constituents of which are selected to permit discrimination of a biological condition, agent or physiological mechanism of action.
  • A “subject” is a cell, tissue, or organism, human or non-human, whether in vivo, ex vivo or in vitro, under observation. When we refer to evaluating the biological condition of a subject based on a sample from the subject, we include using blood or other tissue sample from a human subject to evaluate the human subject's condition; but we also include, for example, using a blood sample itself as the subject to evaluate, for example, the effect of therapy or an agent upon the sample.
  • A “stimulus” includes (i) a monitored physical interaction with a subject, for example ultraviolet A or B, or light therapy for seasonal affective disorder, or treatment of psoriasis with psoralen or treatment of melanoma with embedded radioactive seeds, other radiation exposure, and (ii) any monitored physical, mental, emotional, or spiritual activity or inactivity of a subject.
  • “Therapy” includes all interventions whether biological, chemical, physical, metaphysical, or combination of the foregoing, intended to sustain or alter the monitored biological condition of a subject.
  • The PCT patent application publication number WO 01/25473, published Apr. 12, 2001, entitled “Systems and Methods for Characterizing a Biological Condition or Agent Using Calibrated Gene Expression Profiles,” filed for an invention by inventors herein, and which is herein incorporated by reference, discloses the use of Gene Expression Panels for the evaluation of (i) biological condition (including with respect to health and disease) and (ii) the effect of one or more agents on biological condition (including with respect to health, toxicity, therapeutic treatment and drug interaction).
  • In particular, Gene Expression Panels may be used for measurement of therapeutic efficacy of natural or synthetic compositions or stimuli that may be formulated individually or in combinations or mixtures for a range of targeted biological conditions; prediction of toxicological effects and dose effectiveness of a composition or mixture of compositions for an individual or for a population or set of individuals or for a population of cells; determination of how two or more different agents administered in a single treatment might interact so as to detect any of synergistic, additive, negative, neutral or toxic activity; performing pre-clinical and clinical trials by providing new criteria for pre-selecting subjects according to informative profile data sets for revealing disease status; and conducting preliminary dosage studies for these patients prior to conducting phase 1 or 2 trials. These Gene Expression Panels may be employed with respect to samples derived from subjects in order to evaluate their biological condition.
  • The present invention provides Gene Expression Panels for the evaluation of multiple sclerosis and inflammatory condition related to multiple sclerosis. In addition, the Gene Expression Profiles described herein also provided the evaluation of the effect of one or more agents for the treatment of multiple sclerosis and inflammatory condition related to multiple sclerosis.
  • The Gene Expression Panels (Precision Profile™) are referred to herein as the “Precision Profile™ for Multiple Sclerosis or Inflammatory Conditions Related to Multiple Sclerosis. A Precision Profile™ for Multiple Sclerosis or Inflammatory Conditions Related to Multiple Sclerosis includes one or more genes, e.g., constituents, listed in 1-9. Each gene of the Precision Profile™ for Multiple Sclerosis or Inflammatory Conditions Related to Multiple Sclerosis is referred to herein as a multiple sclerosis associated gene or a multiple sclerosis associated constituent.
  • The evaluation or characterization of multiple sclerosis is defined to be diagnosing multiple sclerosis, assessing the risk of developing multiple sclerosis or assessing the prognosis of a subject with multiple sclerosis. Similarly, the evaluation or characterization of an agent for treatment of multiple sclerosis includes identifying agents suitable for the treatment of multiple sclerosis. The agents can be compounds known to treat multiple sclerosis or compounds that have not been shown to treat multiple sclerosis.
  • Multiple sclerosis and conditions related to multiple sclerosis is evaluated by determining the level of expression (e.g., a quantitative measure) of one or more multiple sclerosis genes. The level of expression is determined by any means known in the art, such as for example quantitative PCR. The measurement is obtained under conditions that are substantially repeatable. Optionally, the qualitative measure of the constituent is compared to a baseline level (e.g. baseline profile set). A baseline level is a level of expression of the constituent in one or more subjects known not to be suffering from multiple sclerosis (e.g., normal, healthy individual(s)). Alternatively, the baseline level is derived from one or more subjects known to be suffering from multiple sclerosis. Optionally, the baseline level is derived from the same subject from which the first measure is derived. For example, the baseline is taken from a subject at different time periods during a course of treatment. Such methods allow for the evaluation of a particular treatment for a selected individual. Comparison can be performed on test (e.g., patient) and reference samples (e.g., baseline) measured concurrently or at temporally distinct times. An example of the latter is the use of compiled expression information, e.g., a gene expression database, which assembles information about expression levels of multiple sclerosis genes.
  • A change in the expression pattern in the patient-derived sample of a multiple sclerosis gene compared to the normal baseline level indicates that the subject is suffering from or is at risk of developing multiple sclerosis. In contrast, when the methods are applied prophylacticly, a similar level compared to the normal control level in the patient-derived sample of a multiple sclerosis gene indicates that the subject is not suffering from or is at risk of developing multiple sclerosis. Whereas, a similarity in the expression pattern in the patient-derived sample of a multiple sclerosis gene compared to the multiple sclerosis baseline level indicates that the subject is suffering from or is at risk of developing multiple sclerosis.
  • Expression of an effective amount of a multiple sclerosis gene also allows for the course of treatment of multiple sclerosis to be monitored. In this method, a biological sample is provided from a subject undergoing treatment, e.g., if desired, biological samples are obtained from the subject at various time points before, during, or after treatment. Expression of an effective amount of a multiple sclerosis gene is then determined and compared to baseline profile. The baseline profile may be taken or derived from one or more individuals who have been exposed to the treatment. Alternatively, the baseline level may be taken or derived from one or more individuals who have not been exposed to the treatment. For example, samples may be collected from subjects who have received initial treatment for multiple sclerosis and subsequent treatment for multiple sclerosis to monitor the progress of the treatment.
  • Differences in the genetic makeup of individuals can result in differences in their relative abilities to metabolize various drugs. Accordingly, the Precision Profile™ for Multiple Sclerosis and Inflammatory Conditions Related to Multiple Sclerosis, disclosed herein, allows for a putative therapeutic or prophylactic to be tested from a selected subject in order to determine if the agent is a suitable for treating or preventing multiple sclerosis in the subject. Additionally, other genes known to be associated with toxicity may be used. By suitable for treatment is meant determining whether the agent will be efficacious, not efficacious, or toxic for a particular individual. By toxic it is meant that the manifestations of one or more adverse effects of a drug when administered therapeutically. For example, a drug is toxic when it disrupts one or more normal physiological pathways.
  • To identify a therapeutic that is appropriate for a specific subject, a test sample from the subject is exposed to a candidate therapeutic agent, and the expression of one or more of multiple sclerosis genes is determined. A subject sample is incubated in the presence of a candidate agent and the pattern of multiple sclerosis gene expression in the test sample is measured and compared to a baseline profile, e.g., a multiple sclerosis baseline profile or a non-multiple sclerosis baseline profile or an index value. The test agent can be any compound or composition. For example, the test agent is a compound known to be useful in the treatment of multiple sclerosis. Alternatively, the test agent is a compound that has not previously been used to treat multiple sclerosis.
  • If the reference sample, e.g., baseline is from a subject that does not have multiple sclerosis a similarity in the pattern of expression of multiple sclerosis genes in the test sample compared to the reference sample indicates that the treatment is efficacious. Whereas a change in the pattern of expression of multiple sclerosis genes in the test sample compared to the reference sample indicates a less favorable clinical outcome or prognosis.
  • By “efficacious” is meant that the treatment leads to a decrease of a sign or symptom of multiple sclerosis in the subject or a change in the pattern of expression of a multiple sclerosis gene in such that the gene expression pattern has an increase in similarity to that of a normal baseline pattern. Assessment of multiple sclerosis is made using standard clinical protocols. Efficacy is determined in association with any known method for diagnosing or treating multiple sclerosis.
  • Agents that are toxic for a specific subject are identified by exposing a test sample from the subject to a candidate agent, and the expression of one or more of multiple sclerosis genes is determined. A subject sample is incubated in the presence of a candidate agent and the pattern of multiple sclerosis gene expression in the test sample is measured and compared to a baseline profile, e.g., a multiple sclerosis baseline profile or a non-multiple sclerosis baseline profile or an index value. The test agent can be any compound or composition. For example, the test agent is a compound known to be useful in the treatment of multiple sclerosis. Alternatively, the test agent is a compound that has not previously been used to treat multiple sclerosis.
  • If the reference sample, e.g., baseline is from a subject in whom the candidate agent is not toxic a similarity in the pattern of expression of multiple sclerosis genes in the test sample compared to the reference sample indicates that the candidate agent is not toxic for the particular subject. Whereas a change in the pattern of expression of multiple sclerosis genes in the test sample compared to the reference sample indicates that the candidate agent is toxic.
  • A Gene Expression Panel (Precision Profile™) is selected in a manner so that quantitative measurement of RNA or protein constituents in the Panel constitutes a measurement of a biological condition of a subject. In one kind of arrangement, a calibrated profile data set is employed. Each member of the calibrated profile data set is a function of (i) a measure of a distinct constituent of a Gene Expression Panel (Precision Profile™) and (ii) a baseline quantity.
  • It has been discovered that valuable and unexpected results may be achieved when the quantitative measurement of constituents is performed under repeatable conditions (within a degree of repeatability of measurement of better than twenty percent, preferably ten percent or better, more preferably five percent or better, and more preferably three percent or better). For the purposes of this description and the following claims, a degree of repeatability of measurement of better than twenty percent may be used as providing measurement conditions that are “substantially repeatable”. In particular, it is desirable that each time a measurement is obtained corresponding to the level of expression of a constituent in a particular sample, substantially the same measurement should result for substantially the same level of expression. In this manner, expression levels for a constituent in a Gene Expression Panel (Precision Profile™) may be meaningfully compared from sample to sample. Even if the expression level measurements for a particular constituent are inaccurate (for example, say, 30% too low), the criterion of repeatability means that all measurements for this constituent, if skewed, will nevertheless be skewed systematically, and therefore measurements of expression level of the constituent may be compared meaningfully. In this fashion valuable information may be obtained and compared concerning expression of the constituent under varied circumstances.
  • In addition to the criterion of repeatability, it is desirable that a second criterion also be satisfied, namely that quantitative measurement of constituents is performed under conditions wherein efficiencies of amplification for all constituents are substantially similar as defined herein. When both of these criteria are satisfied, then measurement of the expression level of one constituent may be meaningfully compared with measurement of the expression level of another constituent in a given sample and from sample to sample.
  • Additional embodiments relate to the use of an index or algorithm resulting from quantitative measurement of constituents, and optionally in addition, derived from either expert analysis or computational biology (a) in the analysis of complex data sets; (b) to control or normalize the influence of uninformative or otherwise minor variances in gene expression values between samples or subjects; (c) to simplify the characterization of a complex data set for comparison to other complex data sets, databases or indices or algorithms derived from complex data sets; (d) to monitor a biological condition of a subject; (e) for measurement of therapeutic efficacy of natural or synthetic compositions or stimuli that may be formulated individually or in combinations or mixtures for a range of targeted biological conditions; (f) for predictions of toxicological effects and dose effectiveness of a composition or mixture of compositions for an individual or for a population or set of individuals or for a population of cells; (g) for determination of how two or more different agents administered in a single treatment might interact so as to detect any of synergistic, additive, negative, neutral of toxic activity (h) for performing pre-clinical and clinical trials by providing new criteria for pre-selecting subjects according to informative profile data sets for revealing disease status and conducting preliminary dosage studies for these patients prior to conducting Phase 1 or 2 trials.
  • Gene expression profiling and the use of index characterization for a particular condition or agent or both may be used to reduce the cost of Phase 3 clinical trials and may be used beyond Phase 3 trials; labeling for approved drugs; selection of suitable medication in a class of medications for a particular patient that is directed to their unique physiology; diagnosing or determining a prognosis of a medical condition or an infection which may precede onset of symptoms or alternatively diagnosing adverse side effects associated with administration of a therapeutic agent; managing the health care of a patient; and quality control for different batches of an agent or a mixture of agents.
  • The Subject
  • The methods disclosed here may be applied to cells of humans, mammals or other organisms without the need for undue experimentation by one of ordinary skill in the art because all cells transcribe RNA and it is known in the art how to extract RNA from all types of cells.
  • A subject can include those who have not been previously diagnosed as having multiple sclerosis or an inflammatory condition related to multiple sclerosis. Alternatively, a subject can also include those who have already been diagnosed as having multiple sclerosis or an inflammatory condition related to multiple sclerosis. Diagnosis of multiple sclerosis is made for example, by clinical data (e.g., episodes of neurologic symptoms characteristic of MS and abnormalities upon physical examination), magnetic resonance imaging of the brain and spine to identify lesions and plaques, testing of cerebral spinal fluid for oligoclonal bands, and measurements of antibodies against myelin proteins (e.g., myelin oligodendrocyte glycoprotein (MOG) and myelin basic protein (MBP).
  • Optionally, the subject has been previously treated with therapeutic agents, or with other therapies and treatment regimens for multiple sclerosis or an inflammatory condition related to multiple sclerosis. A subject can also include those who are suffering from, or at risk of developing multiple sclerosis or an inflammatory condition related to multiple sclerosis, such as those who exhibit known risk factors for multiple sclerosis or an inflammatory condition related to multiple sclerosis. Known risk factors for multiple sclerosis include but are not limited to viral infection, decrease exposure to sunlight, vitamin-D deficiency, chronic infection with spirochetal bacteria and/or Chlamydophila pneumonia, exposure to Epstein-Barr virus, severe stress, and smoking. A subject can include those who are candidates for anti-TNF therapy.
  • Selecting Constituents of a Gene Expression Panel
  • The general approach to selecting constituents of a Gene Expression Panel has been described in PCT application publication number WO 01/25473. A wide range of Gene Expression Panels have been designed and experimentally verified, each panel providing a quantitative measure, of biological condition, that is derived from a sample of blood or other tissue. For each panel, experiments have verified that a Gene Expression Profile using the panel's constituents is informative of a biological condition. (It has also been demonstrated hat in being informative of biological condition, the Gene Expression Profile can be used to used, among other things, to measure the effectiveness of therapy, as well as to provide a target for therapeutic intervention).
  • Tables 1, 2, 3, 4, 5, 6, 7, 8, or 9 listed below, include relevant genes which may be selected for a given Gene Expression Panel, such as the Gene Expression Panels demonstrated herein to be useful in the evaluation of multiple sclerosis and inflammatory condition related to multiple sclerosis.
  • Tables 1-2 were derived from a study of the gene expression patterns described in Example 12 below. Tables 3 and 5-9 were derived from a study of gene expression patterns described in Example 13 below. Table 4 is a panel of 104 genes whose expression is associated with Multiple Sclerosis or Inflammatory Conditions related to Multiple Sclerosis, referred to herein as the Precision Profile™ for Multiple Sclerosis and Inflammatory Genes Related to Multiple Sclerosis. Table 5 shows a ranking of p-values (from most to least significant) of a subset of genes from Table 4. Table 6 describes 2-gene model based on genes from the Precision Profile™ for Multiple sclerosis derived from latent class modeling of the subjects from this study to distinguish between subjects suffering from multiple sclerosis and normal subjects. Two gene models capable of correctly classifying multiple sclerosis-afflicted and/or normal subjects with at least 75% accuracy are indicated. For example, in Table 6, 2-gene model, ITGAL and HLADRA, correctly classifies multiple sclerosis-afflicted subjects with 85.4% accuracy, and normal subjects with 82.9% accuracy.
  • The 2-gene model, CASP9 and HLADRA, correctly classifies multiple sclerosis-afflicted subjects with 78.5% accuracy, and normal subjects with 84.2% accuracy. Table 7 describes 3-gene models based on genes from the Precision Profile™ for Multiple Sclerosis, capable of correctly classifying multiple sclerosis-afflicted and/or normal subjects with at least 75% accuracy are indicated. For example, the three-gene model, ITGAL, HLADRA, and CASP9, correctly classifies multiple sclerosis-afflicted subjects with 85.4% accuracy, and normal subjects with 86.8% accuracy. Table 8 describes a 4-gene model based on genes from the Precision Profile™ for Multiple Sclerosis, capable of correctly classifying multiple sclerosis-afflicted and/or normal subjects with at least 75% accuracy are indicated. For example, the 4-gene model, CASP9, HLADRA, ITGAL, and CCR3, correctly classifies multiple sclerosis-afflicted subjects with 85.4% accuracy, and normal subjects with 83.6% accuracy. Table 9 describes a 5-gene model based on genes from the Precision Profile™ for Multiple Sclerosis, capable of correctly classifying multiple sclerosis-afflicted and/or normal subjects with at least 75% accuracy are indicated. For example, the 4-gene model, CASP9, HLADRA, ITGAL, CCR3, and TGFBR2, correctly classifies multiple sclerosis-afflicted subjects with 86.9% accuracy, and normal subjects with 84.2% accuracy. Table 10 is a panel of 96 genes whose expression is associated with Inflammation referred to herein as the Precision Profile™ for Inflammatory Response.
  • In general, panels may be constructed and experimentally verified by one of ordinary skill in the art in accordance with the principles articulated in the present application.
  • Design of Assays
  • Typically, a sample is run through a panel in replicates of three for each target gene (assay); that is, a sample is divided into aliquots and for each aliquot the concentrations of each constituent in a Gene Expression Panel (Precision Profile™) is measured. From over a total of 900 constituent assays, with each assay conducted in triplicate, an average coefficient of variation was found (standard deviation/average)*100, of less than 2 percent among the normalized ΔCt measurements for each assay (where normalized quantitation of the target mRNA is determined by the difference in threshold cycles between the internal control (e.g., an endogenous marker such as 18S rRNA, or an exogenous marker) and the gene of interest. This is a measure called “intra-assay variability”. Assays have also been conducted on different occasions using the same sample material. This is a measure of “inter-assay variability”. Preferably, the average coefficient of variation of intra-assay variability or inter-assay variability is less than 20%, more preferably less than 10%, more preferably less than 5%, more preferably less than 4%, more preferably less than 3%, more preferably less than 2%, and even more preferably less than 1%.
  • It has been determined that it is valuable to use the quadruplicate or triplicate test results to identify and eliminate data points that are statistical “outliers”; such data points are those that differ by a percentage greater, for example, than 3% of the average of all three or four values. Moreover, if more than one data point in a set of three or four is excluded by this procedure, then all data for the relevant constituent is discarded.
  • Measurement of Gene Expression for a Constituent in the Panel
  • For measuring the amount of a particular RNA in a sample, methods known to one of ordinary skill in the art were used to extract and quantify transcribed RNA from a sample with respect to a constituent of a Gene Expression Panel (Precision Profile™). (See detailed protocols below. Also see PCT application publication number WO 98/24935 herein incorporated by reference for RNA analysis protocols). Briefly, RNA is extracted from a sample such as any tissue, body fluid, cell, or culture medium in which a population of cells of a subject might be growing. For example, cells may be lysed and RNA eluted in a suitable solution in which to conduct a DNAse reaction. Subsequent to RNA extraction, first strand synthesis may be performed using a reverse transcriptase. Gene amplification, more specifically quantitative PCR assays, can then be conducted and the gene of interest calibrated against an internal marker such as 18S rRNA (Hirayama et al., Blood 92, 1998: 46-52). Any other endogenous marker can be used, such as 28S-25S rRNA and 5S rRNA. Samples are measured in multiple replicates, for example, 3 replicates. In an embodiment of the invention, quantitative PCR is performed using amplification, reporting agents and instruments such as those supplied commercially by Applied Biosystems (Foster City, Calif.). Given a defined efficiency of amplification of target transcripts, the point (e.g., cycle number) that signal from amplified target template is detectable may be directly related to the amount of specific message transcript in the measured sample. Similarly, other quantifiable signals such as fluorescence, enzyme activity, disintegrations per minute, absorbance, etc., when correlated to a known concentration of target templates (e.g., a reference standard curve) or normalized to a standard with limited variability can be used to quantify the number of target templates in an unknown sample.
  • Although not limited to amplification methods, quantitative gene expression techniques may utilize amplification of the target transcript. Alternatively or in combination with amplification of the target transcript, quantitation of the reporter signal for an internal marker generated by the exponential increase of amplified product may also be used. Amplification of the target template may be accomplished by isothermic gene amplification strategies or by gene amplification by thermal cycling such as PCR.
  • It is desirable to obtain a definable and reproducible correlation between the amplified target or reporter signal, i.e., internal marker, and the concentration of starting templates. It has been discovered that this objective can be achieved by careful attention to, for example, consistent primer-template ratios and a strict adherence to a narrow permissible level of experimental amplification efficiencies (for example 90.0 to 100%+/−5% relative efficiency, typically 99.8 to 100% relative efficiency). For example, in determining gene expression levels with regard to a single Gene Expression Profile, it is necessary that all constituents of the panels, including endogenous controls, maintain similar amplification efficiencies, as defined herein, to permit accurate and precise relative measurements for each constituent. Amplification efficiencies are regarded as being “substantially similar”, for the purposes of this description and the following claims, if they differ by no more than approximately 10%, preferably by less than approximately 5%, more preferably by less than approximately 3%, and more preferably by less than approximately 1%. Measurement conditions are regarded as being “substantially repeatable, for the purposes of this description and the following claims, if they differ by no more than approximately +/−10% coefficient of variation (CV), preferably by less than approximately +/−5% CV, more preferably +/−2% CV. These constraints should be observed over the entire range of concentration levels to be measured associated with the relevant biological condition. While it is thus necessary for various embodiments herein to satisfy criteria that measurements are achieved under measurement conditions that are substantially repeatable and wherein specificity and efficiencies of amplification for all constituents are substantially similar, nevertheless, it is within the scope of the present invention as claimed herein to achieve such measurement conditions by adjusting assay results that do not satisfy these criteria directly, in such a manner as to compensate for errors, so that the criteria are satisfied after suitable adjustment of assay results.
  • In practice, tests are run to assure that these conditions are satisfied. For example, the design of all primer-probe sets are done in house, experimentation is performed to determine which set gives the best performance. Even though primer-probe design can be enhanced using computer techniques known in the art, and notwithstanding common practice, it has been found that experimental validation is still useful. Moreover, in the course of experimental validation, the selected primer-probe combination is associated with a set of features:
  • The reverse primer should be complementary to the coding DNA strand. In one embodiment, the primer should be located across an intron-exon junction, with not more than four bases of the three-prime end of the reverse primer complementary to the proximal exon. (If more than four bases are complementary, then it would tend to competitively amplify genomic DNA.)
  • In an embodiment of the invention, the primer probe set should amplify cDNA of less than 110 bases in length and should not amplify, or generate fluorescent signal from, genomic DNA or transcripts or cDNA from related but biologically irrelevant loci.
  • A suitable target of the selected primer probe is first strand cDNA, which in one embodiment may be prepared from whole blood as follows:
  • (a) Use of Whole Blood for Ex Vivo Assessment of a Biological Condition Affected by an Agent.
  • Human blood is obtained by venipuncture and prepared for assay by separating samples for baseline, no exogenous stimulus, and pro-cancer stimulus with sufficient volume for at least three time points. Typical pro-cancer stimuli include for example, ionizing radiation, free radicals or DNA damaging agents, and may be used individually or in combination. The aliquots of heparinized, whole blood are mixed with additional test therapeutic compounds and held at 37° C. in an atmosphere of 5% CO2 for 30 minutes. Stimulus is added at varying concentrations, mixed and held loosely capped at 37° C. for the prescribed timecourse. At defined time-points, cells are lysed and RNA extracted by various standard means.
  • Nucleic acids, RNA and or DNA are purified from cells, tissues or fluids of the test population of cells or indicator cell lines. RNA is preferentially obtained from the nucleic acid mix using a variety of standard procedures (or RNA Isolation Strategies, pp. 55-104, in RNA Methodologies, A laboratory guide for isolation and characterization, 2nd edition, 1998, Robert E. Farrell, Jr., Ed., Academic Press), in the present using a filter-based RNA isolation system from Anibion (RNAqueous™, Phenol-free Total RNA Isolation Kit, Catalog #1912, version 9908; Austin, Tex.).
  • In accordance with one procedure, the whole blood assay for Gene Expression Profiles determination was carried out as follows: Human whole blood was drawn into 10 mL Vacutainer tubes with Sodium Heparin. Blood samples were mixed by gently inverting tubes 4-5 times. The blood was used within 10-15 minutes of draw. In the experiments, blood was diluted 2-fold, i.e. per sample per time point, 0.6 mL whole blood +0.6 mL stimulus. The assay medium was prepared and the stimulus added as appropriate.
  • A quantity (0.6 mL) of whole blood was then added into each 12×75 mm polypropylene tube. 0.6 mL of 2×LPS (from E. coli serotype 0127:B8, Sigma#L3880 or serotype 055, Sigma #L4005, 10 ng/mL, subject to change in different lots) into LPS tubes was added. Next, 0.6 mL assay medium was added to the “control” tubes. The caps were closed tightly. The tubes were inverted 2-3 times to mix samples. Caps were loosened to first stop and the tubes incubated at 37° C., 5% CO2 for 6 hours. At 6 hours, samples were gently mixed to resuspend blood cells, and 0.15 mL was removed from each tube (using a micropipettor with barrier tip), and transferred to 0.15 mL of lysis buffer and mixed. Lysed samples were extracted using an ABI 6100 Nucleic Acid Prepstation following the manufacturer's recommended protocol.
  • The samples were then centrifuged for 5 min at 500×g, ambient temperature (IEC centrifuge or equivalent, in microfuge tube adapters in swinging bucket), and as much serum from each tube was removed as possible and discarded. Cell pellets were placed on ice; and RNA extracted as soon as possible using an Ambion RNAqueous kit.
  • (b) Amplification Strategies.
  • Specific RNAs are amplified using message specific primers or random primers. The specific primers are synthesized from data obtained from public databases (e.g., Unigene, National Center for Biotechnology Information, National Library of Medicine, Bethesda, Md.), including information from genomic and cDNA libraries obtained from humans and other animals. Primers are chosen to preferentially amplify from specific RNAs obtained from the test or indicator samples (see, for example, RT PCR, Chapter 15 in RNA Methodologies, A laboratory guide for isolation and characterization, 2nd edition, 1998, Robert E. Farrell, Jr., Ed., Academic Press; or Chapter 22 pp. 143-151, RNA isolation and characterization protocols, Methods in molecular biology, Volume 86, 1998, R. Rapley and D. L. Manning Eds., Human Press, or 14 in statistical refinement of primer design parameters, Chapter 5, pp. 55-72, PCR applications: protocols for functional genomics, M. A. Innis, D. H. Gelfand and J. J. Sninsky, Eds., 1999, Academic Press). Amplifications are carried out in either isothermic conditions or using a thermal cycler (for example, a ABI 9600 or 9700 or 7900 obtained from Applied Biosystems, Foster City, Calif.; see Nucleic acid detection methods, pp. 1-24, in Molecular methods for virus detection, D. L. Wiedbrauk and D. H., Farkas, Eds., 1995, Academic Press). Amplified nucleic acids are detected using fluorescent-tagged detection oligonucleotide probes (see, for example, Taqman™ PCR Reagent Kit, Protocol, part number 402823, Revision A, 1996, Applied Biosystems, Foster City Calif.) that are identified and synthesized from publicly known databases as described for the amplification primers. In the present case, amplified cDNA is detected and quantified using the ABI Prism 7900 Sequence Detection System obtained from Applied Biosystems (Foster City, Calif.). Amounts of specific RNAs contained in the test sample or obtained from the indicator cell lines can be related to the relative quantity of fluorescence observed (see for example, Advances in quantitative PCR technology: 5′ nuclease assays, Y. S. Lie and C. J. Petropolus, Current Opinion in Biotechnology, 1998, 9:43-48, or Rapid thermal cycling and PCR kinetics, pp. 211-229, chapter 14 in PCR applications: protocols for functional genomics, M. A. Innis, D. H. Gelfand and J. J. Sninsky, Eds., 1999, Academic Press).
  • As a particular implementation of the approach described here in detail is a procedure for synthesis of first strand cDNA for use in PCR. This procedure can be used for both whole blood RNA and RNA extracted from cultured cells (i.e. THP-1 cells).
  • Materials
  • 1. Applied Biosystems TAQMAN Reverse Transcription Reagents Kit (P/N 808-0234). Kit Components: 10× TaqMan RT Buffer, 25 mM Magnesium chloride, deoxyNTPs mixture, Random Hexamers, RNase Inhibitor, MultiScribe Reverse Transcriptase (50 U/mL) (2) RNase/DNase free water (DEPC Treated Water from Ambion (P/N 9915G), or equivalent).
  • Methods
  • 1. Place RNase Inhibitor and MultiScribe Reverse Transcriptase on ice immediately. All other reagents can be thawed at room temperature and then placed on ice.
  • 2. Remove RNA samples from −80° C. freezer and thaw at room temperature and then place immediately on ice.
  • 3. Prepare the following cocktail of Reverse Transcriptase Reagents for each 100 mL RT reaction (for multiple samples, prepare extra cocktail to allow for pipetting error):
  • 1 reaction (mL) 11X, e.g. 10 samples (μL)
    10X RT Buffer 10.0 110.0
    25 mM MgCl2 22.0 242.0
    dNTPs 20.0 220.0
    Random Hexamers 5.0 55.0
    RNAse Inhibitor 2.0 22.0
    Reverse Transcriptase 2.5 27.5
    Water 18.5 203.5
    Total: 80.0 880.0 (80 μL per sample)
  • 4. Bring each RNA sample to a total volume of 20 μL in a 1.5 mL microcentrifuge tube (for example, for THP-1 RNA, remove 10 μL RNA and dilute to 20 μL with RNase/DNase free water, for whole blood RNA use 20 μL total RNA) and add 80 μL RT reaction mix from step 5, 2, 3. Mix by pipetting up and down.
  • 5. Incubate sample at room temperature for 10 minutes.
  • 6. Incubate sample at 37° C. for 1 hour.
  • 7. Incubate sample at 90° C. for 10 minutes.
  • 8. Quick spin samples in microcentrifuge.
  • 9. Place sample on ice if doing PCR immediately, otherwise store sample at −20° C. for future use.
  • 10. PCR QC should be run on all RT samples using 18S and β-actin.
  • The use of the primer probe with the first strand cDNA as described above to permit measurement of constituents of a Gene Expression Panel (Precision Profile™) is as follows:
  • Materials
  • 1. 20× Primer/Probe Mix for each gene of interest.
  • 2. 20× Primer/Probe Mix for 18S endogenous control.
  • 3. 2× Taqman Universal PCR Master Mix.
  • 4. cDNA transcribed from RNA extracted from cells.
  • 5. Applied Biosystems 96-Well Optical Reaction Plates.
  • 6. Applied Biosystems Optical Caps, or optical-clear film.
  • 7. Applied Biosystem Prism 7700 or 7900 Sequence Detector.
  • Methods
  • 1. Make stocks of each Primer/Probe mix containing the Primer/Probe for the gene of interest, Primer/Probe for 18S endogenous control, and 2×PCR Master Mix as follows. Make sufficient excess to allow for pipetting error e.g., approximately 10% excess. The following example illustrates a typical set up for one gene with quadruplicate samples testing two conditions (2 plates).
  • 1X (1 well) (μL)
    2X Master Mix 7.5
    20X 18S Primer/Probe Mix 0.75
    20X Gene of interest Primer/Probe Mix 0.75
    Total 9.0
  • 2. Make stocks of cDNA targets by diluting 95 μL of cDNA into 2000 μL of water. The amount of cDNA is adjusted to give Ct values between 10 and 18, typically between 12 and 16.
  • 3. Pipette 9 μL of Primer/Probe mix into the appropriate wells of an Applied Biosystems 384-Well Optical Reaction Plate.
  • 4. Pipette 10 μL of cDNA stock solution into each well of the Applied Biosystems 384-Well Optical Reaction Plate.
  • 5. Seal the plate with Applied Biosystems Optical Caps, or optical-clear film.
  • 6. Analyze the plate on the ABI Prism 7900 Sequence Detector.
  • In another embodiment of the invention, the use of the primer probe with the first strand cDNA as described above to permit measurement of constituents of a Gene Expression Panel (Precision Profile™) is performed using a QPCR assay on Cepheid SmartCycler® and GeneXpert® Instruments as follows:
  • I. To run a QPCR assay in duplicate on the Cepheid SmartCycler® instrument containing three target genes and one reference gene, the following procedure should be followed.
  • A. With 20× Primer/Probe Stocks.
  • Materials
      • 1. SmartMix™-HM lyophilized Master Mix.
      • 2. Molecular grade water.
      • 3. 20× Primer/Probe Mix for the 18S endogenous control gene. The endogenous control gene will be dual labeled with VIC-MGB or equivalent.
      • 4. 20× Primer/Probe Mix for each for target gene one, dual labeled with FAM-BHQ1 or equivalent.
      • 5. 20× Primer/Probe Mix for each for target gene two, dual labeled with Texas Red-BHQ2 or equivalent.
      • 6. 20× Primer/Probe Mix for each for target gene three, dual labeled with Alexa 647-BHQ3 or equivalent.
      • 7. Tris buffer, pH 9.0
      • 8. cDNA transcribed from RNA extracted from sample.
      • 9. SmartCycler® 25 μL tube.
      • 10. Cepheid SmartCycler® instrument.
  • Methods
      • 1. For each cDNA sample to be investigated, add the following to a sterile 650 μL tube.
  • SmartMix ™-HM lyophilized Master Mix 1 bead
    20X 18S Primer/Probe Mix 2.5 μL
    20X Target Gene 1 Primer/Probe Mix 2.5 μL
    20X Target Gene 2 Primer/Probe Mix 2.5 μL
    20X Target Gene 3 Primer/Probe Mix 2.5 μL
    Tris Buffer, pH 9.0 2.5 μL
    Sterile Water 34.5 μL
    Total 47 μL
      • Vortex the mixture for 1 second three times to completely mix the reagents. Briefly centrifuge the tube after vortexing.
      • 2. Dilute the cDNA sample so that a 3 μL addition to the reagent mixture above will give an 18S reference gene CT value between 12 and 16.
      • 3. Add 3 μL of the prepared cDNA sample to the reagent mixture bringing the total volume to 50 μL. Vortex the mixture for 1 second three times to completely mix the reagents. Briefly centrifuge the tube after vortexing.
      • 4. Add 25 μL of the mixture to each of two SmartCycler® tubes, cap the tube and spin for 5 seconds in a microcentrifuge having an adapter for SmartCycler® tubes.
      • 5. Remove the two SmartCycler® tubes from the microcentrifuge and inspect for air bubbles. If bubbles are present, re-spin, otherwise, load the tubes into the SmartCycler® instrument.
      • 6. Run the appropriate QPCR protocol on the SmartCycler®, export the data and analyze the results.
  • B. With Lyophilized SmartBeads™.
  • Materials
      • 1. SmartMix™-HM lyophilized Master Mix.
      • 2. Molecular grade water.
      • 3. SmartBeads™ containing the 18S endogenous control gene dual labeled with VIC-MGB or equivalent, and the three target genes, one dual labeled with FAM-BHQ1 or equivalent, one dual labeled with Texas Red-BHQ2 or equivalent and one dual labeled with Alexa 647-BHQ3 or equivalent.
      • 4. Tris buffer, pH 9.0
      • 5. cDNA transcribed from RNA extracted from sample.
      • 6. SmartCycler® 25 μL tube.
      • 7. Cepheid SmartCycler® instrument.
  • Methods
      • 1. For each cDNA sample to be investigated, add the following to a sterile 650 μL tube.
  • SmartMix ™-HM lyophilized Master Mix 1 bead
    SmartBead ™ containing four primer/probe sets 1 bead
    Tris Buffer, pH 9.0 2.5 μL
    Sterile Water 44.5 μL
    Total 47 μL
      • Vortex the mixture for 1 second three times to completely mix the reagents. Briefly centrifuge the tube after vortexing.
      • 2. Dilute the cDNA sample so that a 3 μL addition to the reagent mixture above will give an 18S reference gene CT value between 12 and 16.
      • 3. Add 3 μL of the prepared cDNA sample to the reagent mixture bringing the total volume to 50 μL. Vortex the mixture for 1 second three times to completely mix the reagents. Briefly centrifuge the tube after vortexing.
      • 4. Add 25 μL of the mixture to each of two SmartCycler® tubes, cap the tube and spin for 5 seconds in a microcentrifuge having an adapter for SmartCycler® tubes.
      • 5. Remove the two SmartCycler® tubes from the microcentrifuge and inspect for air bubbles. If bubbles are present, re-spin, otherwise, load the tubes into the SmartCycler® instrument.
      • 6. Run the appropriate QPCR protocol on the SmartCyclerg, export the data and analyze the results.
        II. To run a QPCR assay on the Cepheid GeneXpert® instrument containing three target genes and one reference gene, the following procedure should be followed. Note that to do duplicates, two self contained cartridges need to be loaded and run on the GeneXpert® instrument.
  • Materials
      • 1. Cepheid GeneXpert® self contained cartridge preloaded with a lyophilized SmartMix™-HM master mix bead and a lyophilized SmartBead™ containing four primer/probe sets.
      • 2. Molecular grade water, containing Tris buffer, pH 9.0.
      • 3. Extraction and purification reagents.
      • 4. Clinical sample (whole blood, RNA, etc.)
      • 5. Cepheid GeneXpert® instrument.
  • Methods
      • 1. Remove appropriate GeneXpert® self contained cartridge from packaging.
      • 2. Fill appropriate chamber of self contained cartridge with molecular grade water with Tris buffer, pH 9.0.
      • 3. Fill appropriate chambers of self contained cartridge with extraction and purification reagents.
      • 4. Load aliquot of clinical sample into appropriate chamber of self contained cartridge.
      • 5. Seal cartridge aand load into GeneXpert® instrument.
      • 6. Run the appropriate extraction and amplification protocol on the GeneXpert® and analyze the resultant data.
  • In other embodiments, any tissue, body fluid, or cell(s) may be used for ex vivo assessment of a biological condition affected by an agent.
  • Methods herein may also be applied using proteins where sensitive quantitative techniques, such as an Enzyme Linked ImmunoSorbent Assay (ELISA) or mass spectroscopy, are available and well-known in the art for measuring the amount of a protein constituent. (see WO 98/24935 herein incorporated by reference).
  • Baseline Profile Data Sets
  • The analyses of samples from single individuals and from large groups of individuals provide a library of profile data sets relating to a particular panel or series of panels. These profile data sets may be stored as records in a library for use as baseline profile data sets. As the term “baseline” suggests, the stored baseline profile data sets serve as comparators for providing a calibrated profile data set that is informative about a biological condition or agent. Baseline profile data sets may be stored in libraries and classified in a number of cross-referential ways. One form of classification may rely on the characteristics of the panels from which the data sets are derived. Another form of classification may be by particular biological condition, e.g., multiple sclerosis. The concept of biological condition encompasses any state in which a cell or population of cells may be found at any one time. This state may reflect geography of samples, sex of subjects or any other discriminator. Some of the discriminators may overlap. The libraries may also be accessed for records associated with a single subject or particular clinical trial. The classification of baseline profile data sets may further be annotated with medical information about a particular subject, a medical condition, and/or a particular agent.
  • The choice of a baseline profile data set for creating a calibrated profile data set is related to the biological condition to be evaluated, monitored, or predicted, as well as, the intended use of the calibrated panel, e.g., as to monitor drug development, quality control or other uses. It may be desirable to access baseline profile data sets from the same subject for whom a first profile data set is obtained or from different subject at varying times, exposures to stimuli, drugs or complex compounds; or may be derived from like or dissimilar populations or sets of subjects. The baseline profile data set may be normal, healthy baseline.
  • The profile data set may arise from the same subject for which the first data set is obtained, where the sample is taken at a separate or similar time, a different or similar site or in a different or similar biological condition. For example, a sample may be taken before stimulation or after stimulation with an exogenous compound or substance, such as before or after therapeutic treatment. The profile data set obtained from the unstimulated sample may serve as a baseline profile data set for the sample taken after stimulation. The baseline data set may also be derived from a library containing profile data sets of a population or set of subjects having some defining characteristic or biological condition. The baseline profile data set may also correspond to some ex vivo or in vitro properties associated with an in vitro cell culture. The resultant calibrated profile data sets may then be stored as a record in a database or library along with or separate from the baseline profile data base and optionally the first profile data set although the first profile data set would normally become incorporated into a baseline profile data set under suitable classification criteria. The remarkable consistency of Gene Expression Profiles associated with a given biological condition makes it valuable to store profile data, which can be used, among other things for normative reference purposes. The normative reference can serve to indicate the degree to which a subject conforms to a given biological condition (healthy or diseased) and, alternatively or in addition, to provide a target for clinical intervention.
  • Selected baseline profile data sets may be also be used as a standard by which to judge manufacturing lots in terms of efficacy, toxicity, etc. Where the effect of a therapeutic agent is being measured, the baseline data set may correspond to Gene Expression Profiles taken before administration of the agent. Where quality control for a newly manufactured product is being determined, the baseline data set may correspond with a gold standard for that product. However, any suitable normalization techniques may be employed. For example, an average baseline profile data set is obtained from authentic material of a naturally grown herbal nutraceutical and compared over time and over different lots in order to demonstrate consistency, or lack of consistency, in lots of compounds prepared for release.
  • Calibrated Data
  • Given the repeatability achieved in measurement of gene expression, described above in connection with “Gene Expression Panels” (Precision Profiles™) and “gene amplification”, it was concluded that where differences occur in measurement under such conditions, the differences are attributable to differences in biological condition. Thus, it has been found that calibrated profile data sets are highly reproducible in samples taken from the same individual under the same conditions. Similarly, it has been found that calibrated profile data sets are reproducible in samples that are repeatedly tested. Also found have been repeated instances wherein calibrated profile data sets obtained when samples from a subject are exposed ex vivo to a compound are comparable to calibrated profile data from a sample that has been exposed to a sample in vivo. Importantly, it has been determined that an indicator cell line treated with an agent can in many cases provide calibrated profile data sets comparable to those obtained from in vivo or ex vivo populations of cells. Moreover, it has been determined that administering a sample from a subject onto indicator cells can provide informative calibrated profile data sets with respect to the biological condition of the subject including the health, disease states, therapeutic interventions, aging or exposure to environmental stimuli or toxins of the subject.
  • Calculation of Calibrated Profile Data Sets and Computational Aids
  • The calibrated profile data set may be expressed in a spreadsheet or represented graphically for example, in a bar chart or tabular form but may also be expressed in a three dimensional representation. The function relating the baseline and profile data may be a ratio expressed as a logarithm. The constituent may be itemized on the x-axis and the logarithmic scale may be on the y-axis. Members of a calibrated data set may be expressed as a positive value representing a relative enhancement of gene expression or as a negative value representing a relative reduction in gene expression with respect to the baseline.
  • Each member of the calibrated profile data set should be reproducible within a range with respect to similar samples taken from the subject under similar conditions. For example, the calibrated profile data sets may be reproducible within one order of magnitude with respect to similar samples taken from the subject under similar conditions. More particularly, the members may be reproducible within 20%, and typically within 10%. In accordance with embodiments of the invention, a pattern of increasing, decreasing and no change in relative gene expression from each of a plurality of gene loci examined in the Gene Expression Panel (Precision Profile™) may be used to prepare a calibrated profile set that is informative with regards to a biological condition, biological efficacy of an agent treatment conditions or for comparison to populations or sets of subjects or samples, or for comparison to populations of cells. Patterns of this nature may be used to identify likely candidates for a drug trial, used alone or in combination with other clinical indicators to be diagnostic or prognostic with respect to a biological condition or may be used to guide the development of a pharmaceutical or nutraceutical through manufacture, testing and marketing.
  • The numerical data obtained from quantitative gene expression and numerical data from calibrated gene expression relative to a baseline profile data set may be stored in databases or digital storage mediums and may be retrieved for purposes including managing patient health care or for conducting clinical trials or for characterizing a drug. The data may be transferred in physical or wireless networks via the World Wide Web, email, or internet access site for example or by hard copy so as to be collected and pooled from distant geographic sites.
  • The method also includes producing a calibrated profile data set for the panel, wherein each member of the calibrated profile data set is a function of a corresponding member of the first profile data set and a corresponding member of a baseline profile data set for the panel, and wherein the baseline profile data set is related to the multiple sclerosis or conditions related to multiple sclerosis to be evaluated, with the calibrated profile data set being a comparison between the first profile data set and the baseline profile data set, thereby providing evaluation of multiple sclerosis or conditions related to multiple sclerosis of the subject.
  • In yet other embodiments, the function is a mathematical function and is other than a simple difference, including a second function of the ratio of the corresponding member of first profile data set to the corresponding member of the baseline profile data set, or a logarithmic function. In such embodiments, the first sample is obtained and the first profile data set quantified at a first location, and the calibrated profile data set is produced using a network to access a database stored on a digital storage medium in a second location, wherein the database may be updated to reflect the first profile data set quantified from the sample. Additionally, using a network may include accessing a global computer network.
  • In an embodiment of the present invention, a descriptive record is stored in a single database or multiple databases where the stored data includes the raw gene expression data (first profile data set) prior to transformation by use of a baseline profile data set, as well as a record of the baseline profile data set used to generate the calibrated profile data set including for example, annotations regarding whether the baseline profile data set is derived from a particular Signature Panel and any other annotation that facilitates interpretation and use of the data.
  • Because the data is in a universal format, data handling may readily be done with a computer. The data is organized so as to provide an output optionally corresponding to a graphical representation of a calibrated data set.
  • For example, a distinct sample derived from a subject being at least one of RNA or protein may be denoted as PI. The first profile data set derived from sample PI is denoted Mj, where Mj is a quantitative measure of a distinct RNA or protein constituent of PI. The record Ri is a ratio of M and P and may be annotated with additional data on the subject relating to, for example, age, diet, ethnicity, gender, geographic location, medical disorder, mental disorder, medication, physical activity, body mass and environmental exposure. Moreover, data handling may further include accessing data from a second condition database which may contain additional medical data not presently held with the calibrated profile data sets. In this context, data access may be via a computer network.
  • The above described data storage on a computer may provide the information in a form that can be accessed by a user. Accordingly, the user may load the information onto a second access site including downloading the information. However, access may be restricted to users having a password or other security device so as to protect the medical records contained within. A feature of this embodiment of the invention is the ability of a user to add new or annotated records to the data set so the records become part of the biological information.
  • The graphical representation of calibrated profile data sets pertaining to a product such as a drug provides an opportunity for standardizing a product by means of the calibrated profile, more particularly a signature profile. The profile may be used as a feature with which to demonstrate relative efficacy, differences in mechanisms of actions, etc. compared to other drugs approved for similar or different uses.
  • The various embodiments of the invention may be also implemented as a computer program product for use with a computer system. The product may include program code for deriving a first profile data set and for producing calibrated profiles. Such implementation may include a series of computer instructions fixed either on a tangible medium, such as a computer readable medium (for example, a diskette, CD-ROM, ROM, or fixed disk), or transmittable to a computer system via a modem or other interface device, such as a communications adapter coupled to a network. The network coupling may be for example, over optical or wired communications lines or via wireless techniques (for example, microwave, infrared or other transmission techniques) or some combination of these. The series of computer instructions preferably embodies all or part of the functionality previously described herein with respect to the system. Those skilled in the art should appreciate that such computer instructions can be written in a number of programming languages for use with many computer architectures or operating systems. Furthermore, such instructions may be stored in any memory device, such as semiconductor, magnetic, optical or other memory devices, and may be transmitted using any communications technology, such as optical, infrared, microwave, or other transmission technologies. It is expected that such a computer program product may be distributed as a removable medium with accompanying printed or electronic documentation (for example, shrink wrapped software), preloaded with a computer system (for example, on system ROM or fixed disk), or distributed from a server or electronic bulletin board over a network (for example, the Internet or World Wide Web). In addition, a computer system is further provided including derivative modules for deriving a first data set and a calibration profile data set.
  • The calibration profile data sets in graphical or tabular form, the associated databases, and the calculated index or derived algorithm, together with information extracted from the panels, the databases, the data sets or the indices or algorithms are commodities that can be sold together or separately for a variety of purposes as described in WO 01/25473.
  • In other embodiments, a clinical indicator may be used to assess the multiple sclerosis or inflammatory conditions related to multiple sclerosis of the relevant set of subjects by interpreting the calibrated profile data set in the context of at least one other clinical indicator, wherein the at least one other clinical indicator is selected from the group consisting of blood chemistry, urinalysis, X-ray or other radiological or metabolic imaging technique, other chemical assays, and physical findings.
  • Index Construction
  • In combination, (i) the remarkable consistency of Gene Expression Profiles with respect to a biological condition across a population or set of subject or samples, or across a population of cells and (ii) the use of procedures that provide substantially reproducible measurement of constituents in a Gene Expression Panel giving rise to a Gene Expression Profile, under measurement conditions wherein specificity and efficiencies of amplification for all constituents of the panel are substantially similar, make possible the use of an index that characterizes a Gene Expression Profile, and which therefore provides a measurement of a biological condition.
  • An index may be constructed using an index function that maps values in a Gene Expression Profile into a single value that is pertinent to the biological condition at hand. The values in a Gene Expression Profile are the amounts of each constituent of the Gene Expression Panel that corresponds to the Gene Expression Profile. These constituent amounts form a profile data set, and the index function generates a single value—the index—from the members of the profile data set.
  • The index function may conveniently be constructed as a linear sum of terms, each term being what we call a “contribution function” of a member of the profile data set. For example, the contribution function may be a constant times a power of a member of the profile data set. So the index function would have the form

  • I=ΣC i M i P(i)),
  • where I is the index, Mi is the value of the member i of the profile data set, Ci is a constant, and P(i) is a power to which Mi is raised, the sum being formed for all integral values of i up to the number of members in the data set. We thus have a linear polynomial expression. The role of the coefficient Ci for a particular gene expression specifies whether a higher ΔCt value for this gene either increases (a positive Ci) or decreases (a lower value) the likelihood of multiple sclerosis, the ΔCt values of all other genes in the expression being held constant.
  • The values Ci and P(i) may be determined in a number of ways, so that the index I is informative of the pertinent biological condition. One way is to apply statistical techniques, such as latent class modeling, to the profile data sets to correlate clinical data or experimentally derived data, or other data pertinent to the biological condition. In this connection, for example, may be employed the software from Statistical Innovations, Belmont, Mass., called Latent Gold®. See the web pages at statisticalinnovations.com/lg/, which are hereby incorporated herein by reference.
  • Alternatively, other simpler modeling techniques may be employed in a manner known in the art. The index function for inflammation may be constructed, for example, in a manner that a greater degree of inflammation (as determined by a profile data set for the Precision Profile™ for Inflammatory Response shown in Table 10) correlates with a large value of the index function. In a simple embodiment, therefore, each P(i) may be +1 or —I, depending on whether the constituent increases or decreases with increasing inflammation. As discussed in further detail below, we have constructed a meaningful inflammation index that is proportional to the expression

  • 1/4{IL1A}+1/4{IL1B}+1/4{TNF}+1/4{INFG}−1/{IL10},
  • where the braces around a constituent designate measurement of such constituent and the constituents are a subset of the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response).
  • Just as a baseline profile data set, discussed above, can be used to provide an appropriate normative reference, and can even be used to create a Calibrated profile data set, as discussed above, based on the normative reference, an index that characterizes a Gene Expression Profile can also be provided with a normative value of the index function used to create the index. This normative value can be determined with respect to a relevant population or set of subjects or samples or to a relevant population of cells, so that the index may be interpreted in relation to the normative value. The relevant population or set of subjects or samples, or relevant population of cells may have in common a property that is at least one of age range, gender, ethnicity, geographic location, nutritional history, medical condition, clinical indicator, medication, physical activity, body mass, and environmental exposure.
  • As an example, the index can be constructed, in relation to a normative Gene Expression Profile for a population or set of healthy subjects, in such a way that a reading of approximately 1 characterizes normative Gene Expression Profiles of healthy subjects. Let us further assume that the biological condition that is the subject of the index is inflammation; a reading of 11n this example thus corresponds to a Gene Expression Profile that matches the norm for healthy subjects. A substantially higher reading then may identify a subject experiencing an inflammatory condition. The use of 1 as identifying a normative value, however, is only one possible choice; another logical choice is to use 0 as identifying the normative value. With this choice, deviations in the index from zero can be indicated in standard deviation units (so that values lying between −1 and +1 encompass 90% of a normally distributed reference population or set of subjects. Since we have found that Gene Expression Profile values (and accordingly constructed indices based on them) tend to be normally distributed, the O-centered index constructed in this manner is highly informative. It therefore facilitates use of the index in diagnosis of disease and setting objectives for treatment. The choice of 0 for the normative value, and the use of standard deviation units, for example, are illustrated in FIG. 17B, discussed below.
  • Still another embodiment is a method of providing an index that is indicative of multiple sclerosis or inflammatory conditions related to multiple sclerosis of a subject based on a first sample from the subject, the first sample providing a source of RNAs, the method comprising deriving from the first sample a profile data set, the profile data set including a plurality of members, each member being a quantitative measure of the amount of a distinct RNA constituent in a panel of constituents selected so that measurement of the constituents is indicative of the presumptive signs of multiple sclerosis, the panel including at least two of the constituents of any of the Tables 1-10. In deriving the profile data set, such measure for each constituent is achieved under measurement conditions that are substantially repeatable, at least one measure from the profile data set is applied to an index function that provides a mapping from at least one measure of the profile data set into one measure of the presumptive signs of multiple sclerosis, so as to produce an index pertinent to the multiple sclerosis or inflammatory conditions related to multiple sclerosis of the subject.
  • As a further embodiment of the invention, we can employ an index function I of the form
  • I = C 0 + i = 1 N C i M i + i = 1 N j = 1 N C ij M i M j ,
  • where Mi and Mj are values respectively of the member i and member j of the profile data set having N members, and Ci and Cij are constants. For example, when Ci=Cij=0, the index function is simply the constant C0. More importantly, when Cij=0, the index function is a linear expression, in a form used for examples herein. Similarly, when Cij=0 only when i≠j, the index function is a simple quadratic expression without cross products Otherwise, the index function is a quadratic with cross products. As discussed in further detail below, a quadratic expression that is constructed as a meaningful identifier of rheumatoid arthritis (RA) is the following:

  • C0+C1{TLR2}+C2{CD4}+C3{NFKB1}+C4{TLR2}{CD4}+C5{TLR2}{NFKB1}+C6{NFKB1}2+C7{TLR2}2+C8{CD4}2,
  • where the constant C0 serves to calibrate this expression to the biological population of interest (such as RA), that is characterized by inflammation.
  • In this embodiment, when the index value associated with a subject equals 0, the odds are 50:50 of the subject's being MS vs normal. More generally, the predicted odds of being MS is [exp(Ii)], and therefore the predicted probability of being MS is [exp(Ii)]/[1+exp((Ii)]. Thus, when the index exceeds 0, the predicted probability that a subject is MS is higher than 0.5, and when it falls below 0, the predicted probability is less than 0.5.
  • The value of C0 may be adjusted to reflect the prior probability of being in this population based on known exogenous risk factors for the subject. In an embodiment where C0 is adjusted as a function of the subject's risk factors, where the subject has prior probability pi of being RA based on such risk factors, the adjustment is made by increasing (decreasing) the unadjusted C0 value by adding to C0 the natural logarithm of the ratio of the prior odds of being RA taking into account the risk factors to the overall prior odds of being RA without taking into account the risk factors.
  • It was determined that the above quadratic expression for RA may be well approximated by a linear expression of the form:

  • D0+D1{TLR2}+D2{CD4}+D3{NFKB1}.
  • Yet another embodiment provides a method of using an index for differentiating a type of pathogen within a class of pathogens of interest in a subject with multiple sclerosis or inflammatory conditions related to multiple sclerosis, based on at least one sample from the subject, the method comprising providing at least one index according to any of the above disclosed embodiments for the subject, comparing the at least one index to at least one normative value of the index, determined with respect to at least one relevant set of subjects to obtain at least one difference, and using the at least one difference between the at least one index and the at least one normative value for the index to differentiate the type of pathogen from the class of pathogen.
  • Kits
  • The invention also includes an MS-detection reagent, i.e., nucleic acids that specifically identify one or more multiple sclerosis or inflammatory condition related to multiple sclerosis nucleic acids (e.g., any gene listed in Tables 1-10; referred to herein as MS-associated genes) by having homologous nucleic acid sequences, such as oligonucleotide sequences, complementary to a portion of the MS-associated genes nucleic acids or antibodies to proteins encoded by the MS-associated genes nucleic acids packaged together in the form of a kit. The oligonucleotides can be fragments of the MS-associated genes genes. For example the oligonucleotides can be 200, 150, 100, 50, 25, 10 or less nucleotides in length. The kit may contain in separate containers a nucleic acid or antibody (either already bound to a solid matrix or packaged separately with reagents for binding them to the matrix), control formulations (positive and/or negative), and/or a detectable label. Instructions (i.e., written, tape, VCR, CD-ROM, etc.) for carrying out the assay may be included in the kit. The assay may for example be in the form of PCR, a Northern hybridization or a sandwich ELISA as known in the art.
  • For example, MS-associated genes detection reagents can be immobilized on a solid matrix such as a porous strip to form at least one MS-associated genes detection site. The measurement or detection region of the porous strip may include a plurality of sites containing a nucleic acid. A test strip may also contain sites for negative and/or positive controls. Alternatively, control sites can be located on a separate strip from the test strip. Optionally, the different detection sites may contain different amounts of immobilized nucleic acids, i.e., a higher amount in the first detection site and lesser amounts in subsequent sites. Upon the addition of test sample, the number of sites displaying a detectable signal provides a quantitative indication of the amount of MS-associated genes present in the sample. The detection sites may be configured in any suitably detectable shape and are typically in the shape of a bar or dot spanning the width of a test strip.
  • Alternatively, multiple sclerosis detection genes can be labeled (e.g., with one or more fluorescent dyes) and immobilized on lyophilized beads to form at least one multiple sclerosis gene detection site. The beads may also contain sites for negative and/or positive controls. Upon addition of the test sample, the number of sites displaying a detectable signal provides a quantitative indication of the amount of multiple sclerosis genes present in the sample.
  • Alternatively, the kit contains a nucleic acid substrate array comprising one or more nucleic acid sequences. The nucleic acids on the array specifically identify one or more nucleic acid sequences represented by MS-associated genes (e.g., any gene listed in Tables 1-10). In various embodiments, the expression of 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 40 or 50 or more of the sequences represented by MS-associated genes can be identified by virtue of binding to the array. The substrate array can be on, i.e., a solid substrate, i.e., a “chip” as described in U.S. Pat. No. 5,744,305. Alternatively, the substrate array can be a solution array, i.e., Luminex, Cyvera, Vitra and Quantum Dots' Mosaic.
  • The skilled artisan can routinely make antibodies, nucleic acid probes, i.e., oligonucleotides, aptamers, siRNAs, antisense oligonucleotides, against any of the MS-associated genes in Tables 1-10.
  • Other Embodiments
  • While the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.
  • EXAMPLES Example 1 Acute Inflammatory Index to Assist in Analysis of Large, Complex Data Sets
  • In one embodiment of the invention the index value or algorithm can be used to reduce a complex data set to a single index value that is informative with respect to the inflammatory state of a subject. This is illustrated in FIGS. 1A and 1B.
  • FIG. 1A is entitled Source Precision Inflammation Profile Tracking of A Subject Results in a Large, Complex Data Set. The figure shows the results of assaying 24 genes from the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response) on eight separate days during the course of optic neuritis in a single male subject. FIG. 1B shows use of an Acute Inflammation Index. The data displayed in FIG. 1A above is shown in this figure after calculation using an index function proportional to the following mathematical expression: (1/4{IL1A}+1/4{IL1B}+1/4{TNF}+1/4{INFG}−1/{IL10}).
  • Example 2 Use of Acute Inflammation Index or Algorithm to Monitor a Biological Condition of a Sample or a Subject
  • The inflammatory state of a subject reveals information about the past progress of the biological condition, future progress, response to treatment, etc. The Acute Inflammation Index may be used to reveal such information about the biological condition of a subject. This is illustrated in FIG. 2.
  • The results of the assay for inflammatory gene expression for each day (shown for 24 genes in each row of FIG. 1A) is displayed as an individual histogram after calculation. The index reveals clear trends in inflammatory status that may correlated with therapeutic intervention (FIG. 2).
  • FIG. 2 is a graphical illustration of the acute inflammation index calculated at 9 different, significant clinical milestones from blood obtained from a single patient treated medically with for optic neuritis. Changes in the index values for the Acute Inflammation Index correlate strongly with the expected effects of therapeutic intervention. Four clinical milestones have been identified on top of the Acute Inflammation Index in this figure including (1) prior to treatment with steroids, (2) treatment with IV solumedrol at 1 gram per day, (3) post-treatment with oral prednisone at 60 mg per day tapered to 10 mg per day and (4) post treatment. The data set is the same as for FIG. 1. The index is proportional to 1/4{IL1A}+1/4{IL1B}+1/4{TNF}+1/4{INFG}−1/{IL10}. As expected, the acute inflammation index falls rapidly with treatment with IV steroid, goes up during less efficacious treatment with oral prednisone and returns to the pre-treatment level after the steroids have been discontinued and metabolized completely.
  • Example 3
  • Use of the acute inflammatory index to set dose, including concentrations and timing, for compounds in development or for compounds to be tested in human and non-human subjects as shown in FIG. 3. The acute inflammation index may be used as a common reference value for therapeutic compounds or interventions without common mechanisms of action. The compound that induces a gene response to a compound as indicated by the index, but fails to ameliorate a known biological conditions may be compared to a different compounds with varying effectiveness in treating the biological condition.
  • FIG. 3 shows the effects of single dose treatment with 800 mg of ibuprofen in a single donor as characterized by the Acute Inflammation Index. 800 mg of over-the-counter ibuprofen were taken by a single subject at Time=0 and Time=48 hr. Gene expression values for the indicated five inflammation-related gene loci were determined as described below at times=2, 4, 6, 48, 50, 56 and 96 hours. As expected the acute inflammation index falls immediately after taking the non-steroidal anti-inflammatory ibuprofen and returns to baseline after 48 hours. A second dose at T=48 follows the same kinetics at the first dose and returns to baseline at the end of the experiment at T=96.
  • Example 4 Use of the Acute Inflammation Index to Characterize Efficacy, Safety and Mode of Physiological Action for an Agent
  • FIG. 4 shows that the calculated acute inflammation index displayed graphically for five different conditions including (A) untreated whole blood; (B) whole blood treated in vitro with DMSO, an non-active carrier compound; (C) otherwise unstimulated whole blood treated in vitro with dexamethasone (0.08 ug/ml); (D) whole blood stimulated in vitro with lipopolysaccharide, a known pro-inflammatory compound, (LPS, 1 ng/ml) and (E) whole blood treated in vitro with LPS (1 ng/ml) and dexamethasone (0.08 ug/ml). Dexamethasone is used as a prescription compound that is commonly used medically as an anti-inflammatory steroid compound. The acute inflammation index is calculated from the experimentally determined gene expression levels of inflammation-related genes expressed in human whole blood obtained from a single patient. Results of mRNA expression are expressed as Ct's in this example, but may be expressed as, e.g., relative fluorescence units, copy number or any other quantifiable, precise and calibrated form, for the genes IL1A, IL1B, TNF, IFNG and IL10. From the gene expression values, the acute inflammation values were determined algebraically according in proportion to the expression 1/4{IL1A}+1/4{IL1B}+1/4{TNF}+1/4{INFG}−1/{IL10}.
  • Example 5 Development and Use of Population Normative Values for Gene Expression Profiles
  • FIGS. 6 and 7 show the arithmetic mean values for gene expression profiles (using the 48 loci of the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response)) obtained from whole blood of two distinct patient populations (patient sets). These patient sets are both normal or undiagnosed. The first patient set, which is identified as Bonfils (the plot points for which are represented by diamonds), is composed of 17 subjects accepted as blood donors at the Bonfils Blood Center in Denver, Colo. The second patient set is 9 donors, for which Gene Expression Profiles were obtained from assays conducted four times over a four-week period. Subjects in this second patient set (plot points for which are represented by squares) were recruited from employees of Source Precision Medicine, Inc., the assignee herein. Gene expression averages for each population were calculated for each of 48 gene loci of the Gene Expression Inflammation Panel. The results for loci 1-24 (sometimes referred to below as the Inflammation 48A loci) are shown in FIG. 6 and for loci 25-48 (sometimes referred to below as the Inflammation 48B loci) are shown in FIG. 7.
  • The consistency between gene expression levels of the two distinct patient sets is dramatic. Both patient sets show gene expressions for each of the 48 loci that are not significantly different from each other. This observation suggests that there is a “normal” expression pattern for human inflammatory genes, that a Gene Expression Profile, using the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response) (or a subset thereof) characterizes that expression pattern, and that a population-normal expression pattern can be used, for example, to guide medical intervention for any biological condition that results in a change from the normal expression pattern.
  • In a similar vein, FIG. 8 shows arithmetic mean values for gene expression profiles
  • (again using the 48 loci of the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response)) also obtained from whole blood of two distinct patient populations (patient sets). One patient set, expression values for which are represented by triangular data points, is 24 normal, undiagnosed subjects (who therefore have no known inflammatory disease). The other patient set, the expression values for which are represented by diamond-shaped data points, is four patients with rheumatoid arthritis and who have failed therapy (who therefore have unstable rheumatoid arthritis).
  • As remarkable as the consistency of data from the two distinct normal patient sets shown in FIGS. 6 and 7 is the systematic divergence of data from the normal and diseased patient sets shown in FIG. 8. In 45 of the shown 48 inflammatory gene loci, subjects with unstable rheumatoid arthritis showed, on average, increased inflammatory gene expression (lower cycle threshold values; Ct), than subjects without disease. The data thus further demonstrate that is possible to identify groups with specific biological conditions using gene expression if the precision and calibration of the underlying assay are carefully designed and controlled according to the teachings herein.
  • FIG. 9, in a manner analogous to FIG. 8, shows the shows arithmetic mean values for gene expression profiles using 24 loci of the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response)) also obtained from whole blood of two distinct patient sets. One patient set, expression values for which are represented by diamond-shaped data points, is 17 normal, undiagnosed subjects (who therefore have no known inflammatory disease) who are blood donors. The other patient set, the expression values for which are represented by square-shaped data points, is 16 subjects, also normal and undiagnosed, who have been monitored over six months, and the averages of these expression values are represented by the square-shaped data points. Thus the cross-sectional gene expression-value averages of a first healthy population match closely the longitudinal gene expression-value averages of a second healthy population, with approximately 7% or less variation in measured expression value on a gene-to-gene basis.
  • FIG. 10 shows the shows gene expression values (using 14 loci of the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response)) obtained from whole blood of 44 normal undiagnosed blood donors (data for 10 subjects of which is shown). Again, the gene expression values for each member of the population (set) are closely matched to those for the entire set, represented visually by the consistent peak heights for each of the gene loci. Other subjects of the set and other gene loci than those depicted here display results that are consistent with those shown here.
  • In consequence of these principles, and in various embodiments of the present invention, population normative values for a Gene Expression Profile can be used in comparative assessment of individual subjects as to biological condition, including both for purposes of health and/or disease. In one embodiment the normative values for a Gene Expression Profile may be used as a baseline in computing a “calibrated profile data set” (as defined at the beginning of this section) for a subject that reveals the deviation of such subject's gene expression from population normative values. Population normative values for a Gene Expression Profile can also be used as baseline values in constructing index functions in accordance with embodiments of the present invention. As a result, for example, an index function can be constructed to reveal not only the extent of an individual's inflammation expression generally but also in relation to normative values.
  • Example 6 Consistency of Expression Values of Constituents in Gene Expression Panels Over Time as Reliable Indicators of Biological Condition
  • FIG. 11 shows the expression levels for each of four genes (of the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response)), of a single subject, assayed monthly over a period of eight months. It can be seen that the expression levels are remarkably consistent over time.
  • FIGS. 12 and 13 similarly show in each case the expression levels for each of 48 genes (of the Inflammation Gene Expression Panel), of distinct single subjects (selected in each case on the basis of feeling well and not taking drugs), assayed, in the case of FIG. 12 weekly over a period of four weeks, and in the case of FIG. 13 monthly over a period of six months. In each case, again the expression levels are remarkably consistent over time, and also similar across individuals.
  • FIG. 14 also shows the effect over time, on inflammatory gene expression in a single human subject, of the administration of an anti-inflammatory steroid, as assayed using the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response). In this case, 24 of 48 loci are displayed. The subject had a baseline blood sample drawn in a PAX RNA isolation tube and then took a single 60 mg dose of prednisone, an anti-inflammatory, prescription steroid. Additional blood samples were drawn at 2 hr and 24 hr post the single oral dose. Results for gene expression are displayed for all three time points, wherein values for the baseline sample are shown as unity on the x-axis. As expected, oral treatment with prednisone resulted in the decreased expression of most of inflammation-related gene loci, as shown by the 2-hour post-administration bar graphs. However, the 24-hour post-administration bar graphs show that, for most of the gene loci having reduced gene expression at 2 hours, there were elevated gene expression levels at 24 hr.
  • Although the baseline in FIG. 14 is based on the gene expression values before drug intervention associated with the single individual tested, we know from the previous example, that healthy individuals tend toward population normative values in a Gene Expression Profile using the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response) (or a subset of it). We conclude from FIG. 14 that in an attempt to return the inflammatory gene expression levels to those demonstrated in FIGS. 6 and 7 (normal or set levels), interference with the normal expression induced a compensatory gene expression response that over-compensated for the drug-induced response, perhaps because the prednisone had been significantly metabolized to inactive forms or eliminated from the subject.
  • FIG. 15, in a manner analogous to FIG. 14, shows the effect over time, via whole blood samples obtained from a human subject, administered a single dose of prednisone, on expression of 5 genes (of the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response)). The samples were taken at the time of administration (t=0) of the prednisone, then at two and 24 hours after such administration. Each whole blood sample was challenged by the addition of 0.1 ng/ml of lipopolysaccharide (a Gram-negative endotoxin) and a gene expression profile of the sample, post-challenge, was determined. It can seen that the two-hour sample shows dramatically reduced gene expression of the 5 loci of the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response), in relation to the expression levels at the time of administration (t=0). At 24 hours post administration, the inhibitory effect of the prednisone is no longer apparent, and at 3 of the 5 loci, gene expression is in fact higher than at t=0, illustrating quantitatively at the molecular level the well-known rebound effect.
  • FIG. 16 also shows the effect over time, on inflammatory gene expression in a single human subject suffering from rheumatoid arthritis, of the administration of a TNF-inhibiting compound, but here the expression is shown in comparison to the cognate locus average previously determined (in connection with FIGS. 6 and 7) for the normal (i.e., undiagnosed, healthy) patient set. As part of a larger international study involving patients with rheumatoid arthritis, the subject was followed over a twelve-week period. The subject was enrolled in the study because of a failure to respond to conservative drug therapy for rheumatoid arthritis and a plan to change therapy and begin immediate treatment with a TNF-inhibiting compound. Blood was drawn from the subject prior to initiation of new therapy (visit 1). After initiation of new therapy, blood was drawn at 4 weeks post change in therapy (visit 2), 8 weeks (visit 3), and 12 weeks (visit 4) following the start of new therapy. Blood was collected in PAX RNA isolation tubes, held at room temperature for two hours and then frozen at −30° C.
  • Frozen samples were shipped to the central laboratory at Source Precision Medicine, the assignee herein, in Boulder, Colo. for determination of expression levels of genes in the 48-gene Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response). The blood samples were thawed and RNA extracted according to the manufacturer's recommended procedure. RNA was converted to cDNA and the level of expression of the 48 inflammatory genes was determined. Expression results are shown for 11 of the 48 loci in FIG. 16. When the expression results for the 111 loci are compared from visit one to a population average of normal blood donors from the United States, the subject shows considerable difference. Similarly, gene expression levels at each of the subsequent physician visits for each locus are compared to the same normal average value. Data from visits 2, 3 and 4 document the effect of the change in therapy. In each visit following the change in the therapy, the level of inflammatory gene expression for 10 of the 111 loci is closer to the cognate locus average previously determined for the normal (i.e., undiagnosed, healthy) patient set.
  • FIG. 17A further illustrates the consistency of inflammatory gene expression, illustrated here with respect to 7 loci of (of the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response)), in a set of 44 normal, undiagnosed blood donors. For each individual locus is shown the range of values lying within ±2 standard deviations of the mean expression value, which corresponds to 95% of a normally distributed population. Notwithstanding the great width of the confidence interval (95%), the measured gene expression value (ΔCT)—remarkably—still lies within 10% of the mean, regardless of the expression level involved. As described in further detail below, for a given biological condition an index can be constructed to provide a measurement of the condition. This is possible as a result of the conjunction of two circumstances: (i) there is a remarkable consistency of Gene Expression Profiles with respect to a biological condition across a population and (ii) there can be employed procedures that provide substantially reproducible measurement of constituents in a Gene Expression Panel giving rise to a Gene Expression Profile, under measurement conditions wherein specificity and efficiencies of amplification for all constituents of the panel are substantially similar and which therefore provides a measurement of a biological condition. Accordingly, a function of the expression values of representative constituent loci of FIG. 17A is here used to generate an inflammation index value, which is normalized so that a reading of 1 corresponds to constituent expression values of healthy subjects, as shown in the right-hand portion of FIG. 17A.
  • In FIG. 17B, an inflammation index value was determined for each member of a set of 42 normal undiagnosed blood donors, and the resulting distribution of index values, shown in the figure, can be seen to approximate closely a normal distribution, notwithstanding the relatively small subject set size. The values of the index are shown relative to a O-based median, with deviations from the median calibrated in standard deviation units. Thus 90% of the subject set lies within +1 and −1 of a 0 value. We have constructed various indices, which exhibit similar behavior.
  • FIG. 17C illustrates the use of the same index as FIG. 17B, where the inflammation median for a normal population of subjects has been set to zero and both normal and diseased subjects are plotted in standard deviation units relative to that median. An inflammation index value was determined for each member of a normal, undiagnosed population of 70 individuals (black bars). The resulting distribution of index values, shown in FIG. 17C, can be seen to approximate closely a normal distribution. Similarly, index values were calculated for individuals from two diseased population groups, (1) rheumatoid arthritis patients treated with methotrexate (MTX) who are about to change therapy to more efficacious drugs (e.g., TNF inhibitors) (hatched bars), and (2) rheumatoid arthritis patients treated with disease modifying anti-rheumatoid drugs (DMARDS) other than MTX, who are about to change therapy to more efficacious drugs (e.g., MTX). Both populations of subjects present index values that are skewed upward (demonstrating increased inflammation) in comparison to the normal distribution. This figure thus illustrates the utility of an index to derived from Gene Expression Profile data to evaluate disease status and to provide an objective and quantifiable treatment objective. When these two populations of subjects were treated appropriately, index values from both populations returned to a more normal distribution (data not shown here).
  • FIG. 18 plots, in a fashion similar to that of FIG. 17A, Gene Expression Profiles, for the same 7 loci as in FIG. 17A, two different 6-subject populations of rheumatoid arthritis patients. One population (called “stable” in the figure) is of patients who have responded well to treatment and the other population (called “unstable” in the figure) is of patients who have not responded well to treatment and whose therapy is scheduled for change. It can be seen that the expression values for the stable patient population, lie within the range of the 95% confidence interval, whereas the expression values for the unstable patient population for 5 of the 7 loci are outside and above this range. The right-hand portion of the figure shows an average inflammation index of 9.3 for the unstable population and an average inflammation index of 1.8 for the stable population, compared to 1 for a normal undiagnosed population of patients. The index thus provides a measure of the extent of the underlying inflammatory condition, in this case, rheumatoid arthritis. Hence the index, besides providing a measure of biological condition, can be used to measure the effectiveness of therapy as well as to provide a target for therapeutic intervention.
  • FIG. 19 thus illustrates use of the inflammation index for assessment of a single subject suffering from rheumatoid arthritis, who has not responded well to traditional therapy with methotrexate. The inflammation index for this subject is shown on the far right at start of a new therapy (a TNF inhibitor), and then, moving leftward, successively, 2 weeks, 6 weeks, and 12 weeks thereafter. The index can be seen moving towards normal, consistent with physician observation of the patient as responding to the new treatment.
  • FIG. 20 similarly illustrates use of the inflammation index for assessment of three subjects suffering from rheumatoid arthritis, who have not responded well to traditional therapy with methotrexate, at the beginning of new treatment (also with a TNF inhibitor), and 2 weeks and 6 weeks thereafter. The index in each case can again be seen moving generally towards normal, consistent with physician observation of the patients as responding to the new treatment.
  • Each of FIGS. 21-23 shows the inflammation index for an international group of subjects, suffering from rheumatoid arthritis, each of whom has been characterized as stable (that is, not anticipated to be subjected to a change in therapy) by the subjects' treating physician. FIG. 21 shows the index for each of 10 patients in the group being treated with methotrexate, which known to alleviate symptoms without addressing the underlying disease. FIG. 22 shows the index for each of 10 patients in the group being treated with Enbrel (an TNF inhibitor), and FIG. 23 shows the index for each 10 patients being treated with Remicade (another TNF inhibitor). It can be seen that the inflammation index for each of the patients in FIG. 21 is elevated compared to normal, whereas in FIG. 22, the patients being treated with Enbrel as a class have an inflammation index that comes much closer to normal (80% in the normal range). In FIG. 23, it can be seen that, while all but one of the patients being treated with Remicade have an inflammation index at or below normal, two of the patients have an abnormally low inflammation index, suggesting an immunosuppressive response to this drug. (Indeed, studies have shown that Remicade has been associated with serious infections in some subjects, and here the immunosuppressive effect is quantified.) Also in FIG. 23, one subject has an inflammation index that is significantly above the normal range. This subject in fact was also on a regimen of an anti-inflammation steroid (prednisone) that was being tapered; within approximately one week after the inflammation index was sampled, the subject experienced a significant flare of clinical symptoms.
  • Remarkably, these examples show a measurement, derived from the assay of blood taken from a subject, pertinent to the subject's arthritic condition. Given that the measurement pertains to the extent of inflammation, it can be expected that other inflammation-based conditions, including, for example, cardiovascular disease, may be monitored in a similar fashion.
  • FIG. 24 illustrates use of the inflammation index for assessment of a single subject suffering from inflammatory bowel disease, for whom treatment with Remicade was initiated in three doses. The graphs show the inflammation index just prior to first treatment, and then 24 hours after the first treatment; the index has returned to the normal range. The index was elevated just prior to the second dose, but in the normal range prior to the third dose. Again, the index, besides providing a measure of biological condition, is here used to measure the effectiveness of therapy (Remicade), as well as to provide a target for therapeutic intervention in terms of both dose and schedule.
  • FIG. 25 shows Gene Expression Profiles with respect to 24 loci (of the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response)) for whole blood treated with Ibuprofen in vitro in relation to other non-steroidal anti-inflammatory drugs (NSAIDs). The profile for Ibuprofen is in front. It can be seen that all of the NSAIDs, including Ibuprofen share a substantially similar profile, in that the patterns of gene expression across the loci are similar. Notwithstanding these similarities, each individual drug has its own distinctive signature.
  • FIG. 26 illustrates how the effects of two competing anti-inflammatory compounds can be compared objectively, quantitatively, precisely, and reproducibly. In this example, expression of each of a panel of two genes (of the Inflammation Gene Expression Panel (Precision Profile™ for Inflammatory Response)) is measured for varying doses (0.08-250 μg/ml) of each drug in vitro in whole blood. The market leader drug shows a complex relationship between dose and inflammatory gene response. Paradoxically, as the dose is increased, gene expression for both loci initially drops and then increases in the case the case of the market leader. For the other compound, a more consistent response results, so that as the dose is increased, the gene expression for both loci decreases more consistently.
  • FIGS. 27 through 41 illustrate the use of gene expression panels in early identification and monitoring of infectious disease. These figures plot the response, in expression products of the genes indicated, in whole blood, to the administration of various infectious agents or products associated with infectious agents. In each figure, the gene expression levels are “calibrated”, as that term is defined herein, in relation to baseline expression levels determined with respect to the whole blood prior to administration of the relevant infectious agent. In this respect the figures are similar in nature to various figures of our below-referenced patent application WO 01/25473 (for example, FIG. 15 therein). The concentration change is shown ratiometrically, and the baseline level of 1 for a particular gene locus corresponds to an expression level for such locus that is the same, monitored at the relevant time after addition of the infectious agent or other stimulus, as the expression level before addition of the stimulus. Ratiometric changes in concentration are plotted on a logarithmic scale. Bars below the unity line represent decreases in concentration and bars above the unity line represent increases in concentration, the magnitude of each bar indicating the magnitude of the ratio of the change. We have shown in WO 01/25473 and other experiments that, under appropriate conditions, Gene Expression Profiles derived in vitro by exposing whole blood to a stimulus can be representative of Gene Expression Profiles derived in vivo with exposure to a corresponding stimulus.
  • FIG. 27 uses a novel bacterial Gene Expression Panel of 24 genes, developed to discriminate various bacterial conditions in a host biological system. Two different stimuli are employed: lipotechoic acid (LTA), a gram positive cell wall constituent, and lipopolysaccharide (LPS), a gram negative cell wall constituent. The final concentration immediately after administration of the stimulus was 100 ng/mL, and the ratiometric changes in expression, in relation to pre-administration levels, were monitored for each stimulus 2 and 6 hours after administration. It can be seen that differential expression can be observed as early as two hours after administration, for example, in the IFNα2 locus, as well as others, permitting discrimination in response between gram positive and gram negative bacteria.
  • FIG. 28 shows differential expression for a single locus, IFNG, to LTA derived from three distinct sources: S. pyrogenes, B. subtilis, and S. aureus. Each stimulus was administered to achieve a concentration of 100 ng/mL, and the response was monitored at 1, 2, 4, 6, and 24 hours after administration. The results suggest that Gene Expression Profiles can be used to distinguish among different infectious agents, here different species of gram positive bacteria.
  • FIGS. 29 and 30 show the response of the Inflammation 48A and 48B loci respectively (discussed above in connection with FIGS. 6 and 7 respectively) in whole blood to administration of a stimulus of S. aureus and of a stimulus of E. coli (in the indicated concentrations, just after administration, of 107 and 106 CFU/mL respectively), monitored 2 hours after administration in relation to the pre-administration baseline. The figures show that many of the loci respond to the presence of the bacterial infection within two hours after infection.
  • FIGS. 31 and 32 correspond to FIGS. 29 and 30 respectively and are similar to them, with the exception that the monitoring here occurs 6 hours after administration. More of the loci are responsive to the presence of infection. Various loci, such as IL2, show expression levels that discriminate between the two infectious agents.
  • FIG. 33 shows the response of the Inflammation 48A loci to the administration of a stimulus of E. Coli (again in the concentration just after administration of 10 CFU/mL) and to the administration of a stimulus of an E. coli filtrate containing E. coli bacteria by products but lacking E. coli bacteria. The responses were monitored at 2, 6, and 24 hours after administration. It can be seen, for example, that the responses over time of loci IL1B, IL18 and CSF3 to E. coli and to E. coli filtrate are different.
  • FIG. 34 is similar to FIG. 33, but here the compared responses are to stimuli from E. coli filtrate alone and from E. coli filtrate to which has been added polymyxin B, an antibiotic known to bind to lipopolysaccharide (LPS). An examination of the response of IL1B, for example, shows that presence of polymyxin B did not affect the response of the locus to E. coli filtrate, thereby indicating that LPS does not appear to be a factor in the response of IL1B to E. coli filtrate.
  • FIG. 35 illustrates the responses of the Inflammation 48A loci over time of whole blood to a stimulus of S. aureus (with a concentration just after administration of 10 CFU/mL) monitored at 2, 6, and 24 hours after administration. It can be seen that response over time can involve both direction and magnitude of change in expression. (See for example, IL5 and IL18.)
  • FIGS. 36 and 37 show the responses, of the Inflammation 48A and 48B loci respectively, monitored at 6 hours to stimuli from E. coli (at concentrations of 106 and 102 CFU/mL immediately after administration) and from S. aureus (at concentrations of 107 and 102 CFU/mL immediately after administration). It can be seen, among other things, that in various loci, such as B7 (FIG. 36), TACI, PLA2G7, and C1QA (FIG. 37), E. coli produces a much more pronounced response than S. aureus. The data suggest strongly that Gene Expression Profiles can be used to identify with high sensitivity the presence of gram negative bacteria and to discriminate against gram positive bacteria.
  • FIGS. 38 and 39 show the responses, of the Inflammation 48B and 48A loci respectively, monitored 2, 6, and 24 hours after administration, to stimuli of high concentrations of S. aureus and E. coli respectively (at respective concentrations of 107 and 106 CFU/mL immediately after administration). The responses over time at many loci involve changes in magnitude and direction. FIG. 40 is similar to FIG. 39, but shows the responses of the Inflammation 48B loci.
  • FIG. 41 similarly shows the responses of the Inflammation 48A loci monitored at 24 hours after administration to stimuli high concentrations of S. aureus and E. coli respectively (at respective concentrations of 107 and 106 CFU/mL immediately after administration). As in the case of FIGS. 20 and 21, responses at some loci, such as GRO1 and GRO2, discriminate between type of infection.
  • FIG. 42 illustrates application of a statistical T-test to identify potential members of a signature gene expression panel that is capable of distinguishing between normal subjects and subjects suffering from unstable rheumatoid arthritis. The grayed boxes show genes that are individually highly effective (t test P values noted in the box to the right in each case) in distinguishing between the two sets of subjects, and thus indicative of potential members of a signature gene expression panel for rheumatoid arthritis.
  • FIG. 43 illustrates, for a panel of 17 genes, the expression levels for 8 patients presumed to have bacteremia. The data are suggestive of the prospect that patients with bacteremia have a characteristic pattern of gene expression.
  • FIG. 44 illustrates application of a statistical T-test to identify potential members of a signature gene expression panel that is capable of distinguishing between normal subjects and subjects suffering from bacteremia. The grayed boxes show genes that are individually highly effective (t test P values noted in the box to the right in each case) in distinguishing between the two sets of subjects, and thus indicative of potential members of a signature gene expression panel for bacteremia.
  • FIG. 45 illustrates application of an algorithm (shown in the figure), providing an index pertinent to rheumatoid arthritis (RA) as applied respectively to normal subjects, RA patients, and bacteremia patients. The index easily distinguishes RA subjects from both normal subjects and bacteremia subjects.
  • FIG. 46 illustrates application of an algorithm (shown in the figure), providing an index pertinent to bacteremia as applied respectively to normal subjects, rheumatoid arthritis patients, and bacteremia patients. The index easily distinguishes bacteremia subjects from both normal subjects and rheumatoid arthritis subjects.
  • Example 7 High Precision Gene Expression Analysis of an Individual with RRMS
  • A female subject with a long, documented history of relapsing, remitting multiple sclerosis (RRMS) sought medical attention from a neurologist for increasing lower trunk muscle weakness (Visit 1). Blood was drawn for several assays and the subject was given 5 mg prednisone at that visit. Increasing weakness and spreading of the involvement caused subject to return to the neurologist 6 days later. Blood was drawn and the subject was started on 100 mg prednisone and tapered to 5 mg over one week. The subject reported that her muscle weakness subsided rapidly. The subject was seen for a routine visit (visit 3) more than 2 months later. The patient reported no signs of illness at that visit.
  • Results of high precision gene expression analysis are shown below in FIG. 47. The “y” axis reports the gene expression level in standard deviation units compared to the Source Precision Medicine Normal Reference Population Value for that gene locus at dates May 22, 2002 (before prednisone treatment), May 28, 2002 (after 5 mg treatment on May 22) and Jul. 15, 2002 (after 100 mg prednisone treatment on May 28, tapering to 5 mg within one week). Expression Results for several genes from the 72 gene locus Multiple Sclerosis Precision Profile (shown in Tables 1B and 2, which were selected from gene panel shown in Table 4) are shown along the “x” axis. Some gene loci, for example IL18; IL1B; MMP9; PTGS2, reflect the severity of the signs while other loci, for example IL10, show effects induced by the steroid treatment. Other loci reflect the non-relapsing TIMP1; TNF; HMOX1.
  • Example 8 Experimental Design for Identification and Selection of Diagnostic and Prognostic Markers for Evaluating Multiple Sclerosis (Before, During, and After Flare)
  • Samples of whole blood from patients with relapsing remitting multiple sclerosis (RRMS) were collected while their disease is clinically inactive. Additional samples were collected during a clinical exacerbation of the MS (or attack). Levels of gene expression of mediators of inflammatory processes are examined before, during, and after the episode, whether or not anti-inflammatory treatment is employed. The post-attack samples were then compared to samples obtained at baseline and those obtained during the exacerbation, prior to initiation of any anti-inflammatory medication. The results of this study were compared to a database of normal subjects to identify and select diagnostic and prognostic markers of MS activity useful in the Gene Expression Panels for characterizing and evaluating MS according to the invention. Selected markers were tested in additional trials in patients known to have MS, and those suspected of having MS. By using genes selected to be especially probative in characterizing MS and inflammation related to MS, such conditions are identified in patients using the herein-described gene expression profile techniques and methods of characterizing multiple sclerosis or inflammatory conditions related to multiple sclerosis in a subject based on a sample from the subject. These data demonstrate the ability to evaluate, diagnose and characterize MS and inflammatory conditions related to MS in a subject, or population of subjects.
  • In this system, RRMS subjects experiencing a clinical exacerbation showed altered inflammatory-immune response gene expression compared to RRMS patients during remission and healthy subjects. Additionally, gene expression changes are evident in patients who have exacerbations coincident with initiation and completion of treatment.
  • This system thus provides a gene expression assay system for monitoring MS patients that is predictive of disease progression and treatment responsiveness. In using this system, gene expression profile data sets were determined and prepared from inflammation and immune-response related genes (mRNA and protein) in whole blood samples taken from RRMS patients before, during and after clinical exacerbation. Samples taken during an exacerbation were collected prior to treatment for the attack. Gene expression results were then correlated with relevant clinical indices as described.
  • In addition, the observed data in the gene expression profile data sets was compared to reference profile data sets determined from samples from undiagnosed healthy subjects (normals), gene expression profiles for other chronic immune-related genes, and to profile data sets determined for the individual patients during and after the attack. If desired, a subset of the selected identified genes is coupled with appropriate predictive biomedical algorithms for use in predicting and monitoring RRMS disease activity.
  • A study was conducted with 14 patients. Patients were required to have an existing diagnosis of RRMS and be clinically stable for at least thirty days prior to enrollment. Some patients were using disease-modifying medication (Interferon or Glatirimer Acetate). All patients are sampled at baseline, defined as a time when the subject is not currently experiencing an attack (see inclusion criteria). Those who experienced significant neurological symptoms, suggestive of a clinical exacerbation, were sampled prior to any treatment for the attack. If the patient was found to have a clinical exacerbation, then a repeat sample is obtained four weeks later, regardless of whether the patient receives steroids or other treatment for the exacerbation.
  • A clinical exacerbation is defined as the appearance of a new symptom or worsening/reoccurrence of an old symptom, attributed to RRMS, lasting at least 24 hours in the absence of fever, and preceded by stability or improvement for at least 30 days.
  • Each subject was asked to provide a complete medical history including any existing laboratory test results (i.e. MRI, EDSS scores, blood chemistry, hematology, etc) relevant to the patient's MS contained within the patient's medical records. Additional test results (ordered while the subject is enrolled in the study) relating to the treatment of the patient's MS were collected and correlated with gene expression analysis.
  • Subjects who participated in the study met all of the following criteria:
      • 1. Male or Female subjects at least 18 years old with clinically documented active Relapsing-Remitting MS (RRMS) characterized by clearly defined acute attacks followed by full or partial recovery to the pre-existing level of disability, and by a lack of disease progression in the periods between attacks.
      • 2. Subjects are clinically stable for a minimum of 30 days or for a time period determined at the clinician's discretion.
      • 3. Patients are stable (at least three-months) on Interferon therapy or Glatiramer Acetate or are therapy naïve or without the above mentioned therapy for 4 weeks.
      • 4. Subjects must be willing to give written informed consent and to comply with the requirements of the study protocol.
  • Subjects are excluded from the study if they meet any of the following criteria:
      • 1. Primary progressive multiple sclerosis (PPMS).
      • 2. Immunosuppressive therapy (such as azathioprine and MTX) within three months of study participation. Subjects having prior treatment with cyclophosphamide, total lymphoid irradiation, mitoxantrone, cladribine, or bone marrow transplantation, regardless of duration, are also excluded.
      • 3. Corticosteroid therapy within four weeks of participation of the study.
      • 4. Use of any investigational drug with the intent to treat MS or the symptoms of MS within six months of participation in this trial (agents for the symptomatic treatment of MS, e.g., 4-aminopyridine <4-AP>, may be allowed following discussion with Clinician).
      • 5. Infection or risk factors for severe infections, including: excessive immunosuppression including human immunodeficiency virus (HIV) infection; severe, recurrent, or persistent infections (such as Hepatitis B or C, recurrent urinary tract infection or pneumonia); evidence of current inactive or active tuberculosis (TB) infection including recent exposure to M. tuberculosis (converters to a positive purified protein derivative); subjects with a positive PPD or a chest X-ray suggestive of prior TB infection; active Lyme disease; active syphilis; any significant infection requiring hospitalization or IV antibiotics in the month prior to study participation; infection requiring treatment with antibiotics in the two weeks prior to study participation.
      • 6. Any of the following risk factors for development of malignancy: history of lymphoma or leukemia; treatment of cutaneous squamous-cell or basal cell carcinoma within 2 years of enrollment into the study; other malignancy within 5 years; disease associated with an increased risk of malignancy.
      • 7. Other diseases (in addition to MS) that produce neurological manifestations, such as amyotrophic lateral sclerosis, Guillain-Barre syndrome, muscular dystrophy, etc.)
      • 8. Pregnant or lactating females.
    Example 9 Experimental Design for Identification and Selection of Diagnostic and Prognostic Markers for Evaluating Multiple Sclerosis (pre and post Therapy)
  • These studies were designed to identify possible markers of disease activity in multiple sclerosis (MS) to aid in selecting genes for particular Gene Expression Panels. Similar to the previously-described example, the results of this study were compared to a database of gene expression profile data sets determined and obtained from samples from healthy subjects, and the results were used to identify possible markers of MS activity to be used in Gene Expression Panels for characterizing and evaluating MS according to described embodiments. Selected markers were then tested in additional trials to assess their predictive value.
  • Eleven subjects were used in this study. Initially, a smaller number of patients were evaluated, and gene expression profile data sets were determined for these patients and the expression profiles of selected inflammatory markers were assessed. Additional subjects were added to the study after preliminary evidence for particular disease activity markers is obtained so that a larger or more particular panel of genes is selected for determining profile data sets for the full number of subjects in the study.
  • Patients who were not receiving disease-modifying therapy such as interferon were of particular interest but inclusion of patients receiving such therapy was also useful. Patients were asked to give blood at two timepoints—first at enrollment and then again at 3-12 months after enrollment. Clinical data relating to present and history of disease activity, concomitant medications, lab and MRI results, as well as general health assessment questionnaires were also collected.
  • Subjects who participated in the study met the following criteria:
      • 1. Patients having MS that meets the criteria of McDonald et al. Ann Neurol. 2001 July; 50(1):121-7.
      • 2. Patients with clinically active disease as shown by ≧1 exacerbation in previous 12 months.
      • 3. Patients not in acute relapse
      • 4. Patients willing to provide up to 10 ml of blood at up to 3 time points
  • In addition, patients with known hepatitis or HIV infection were not eligible. The enrollment samples from suitable subjects were collected prior to the patient receiving any disease modifying therapy. The later samples were collected 3-12 months after the patients start therapy. Preliminary data suggests that gene expression can used to track drug response and that only a plurality or several genetic markers is required to identify MS in a population of samples.
  • Example 10 Experimental Design for Identification and Selection of Diagnostic and Prognostic Markers for Evaluating Multiple Sclerosis (Dosing Safety and Response)
  • Theses studies were designed to identify biomarkers for use in a specific Gene Expression Panel for MS, wherein the genes/biomarkers were selected to evaluate dosing and safety of a new compound developed for treating MS, and to track drug response. Specifically a multi-center, randomized, double blind, placebo-controlled trial was used to evaluate a new drug therapy in patients with multiple sclerosis.
  • Thirty subjects were enrolled in this study. Only patients who exhibited stable MS for three months prior to the study were selected for the trial. Stable disease is defined as the absence of progression and relapse. Subjects enrolled in this study had been removed from disease modifying therapy for at least 1 month. A subject's clinical status was monitored throughout the study by MRI and hematology and blood chemistries.
  • Throughout the study patients received all medications necessary for management of their MS, including high-dose corticosteroids for management of relapses and introduction of standard treatments for MS. Initiation of such treatments will confound assessment of the trial's endpoints. Consequently, patients who required such treatment were removed from the new drug therapy phase of the trial but were continued to be followed for safety, immune response, and gene expression.
  • Blood samples for gene expression analysis were collected at screening/baseline (prior to initiation of drug), several times during the treatment phase and several times during follow-up (post-treatment phase). Gene expression results were compared within subjects, between subjects, and to Source Precision Medicine profile data sets determined to be what are termed “Normals”—i.e., a baseline profile dataset determined for a population of healthy (undiagnosed) individuals who do not have MS or other inflammatory conditions, disease, infections. The results were also evaluated to compare and contrast gene expression between different timepoints. This study was used to track individual and population response to the drug, and to correlate clinical symptoms (i.e. disease progression, disease remittance, adverse events) with gene expression.
  • Baseline samples from a subset of patients were analyzed. The preliminary data from the baseline samples suggest that that only a plurality of or optionally several specific genetic markers is required to identify MS across a population of samples. The study was also used to track drug response and clinical endpoints.
  • Example 11 Experimental Design for Identification and Selection of Diagnostic and Prognostic Markers for Evaluating Multiple Sclerosis (Testing Treatment)
  • Theses studies were designed a study for testing a new experimental treatment for MS. The study enrolled 200 MS subjects in a Phase 2, multi-center, randomized, double-blind, parallel group, placebo-controlled, dose finding, safety, tolerability, and efficacy study. Samples for gene expression were collected at baseline and at several timepoints during the study. Samples were compared between subjects, within individual subjects, and to Source Precision Medicine profile data sets determined to be what are termed “Normals”—i.e., a baseline profile dataset determined for a population of healthy (undiagnosed) individuals who do not have MS or other inflammatory conditions, disease, infections. The gene expression profile data sets were then assessed for their ability to track individual response to therapy, for identifying a subset of genes that exhibit altered gene expression in MS and/or are affected by the drug treatment. Clinical data collected during the study include: MRIs, disease progression tests (EDSS, MSFC, ambulation tests, auditory testing, dexterity testing), medical history, concomitant medications, adverse events, physical exam, hematology and chemistry labs, urinalysis, and immunologic testing.
  • Subjects enrolled in the study were asked to discontinue any MS disease modifying therapies they may be using for their disease for at least 3 months prior to dosing with the study drug or drugs.
  • Example 12 Clinical Data Analyzed with Latent Class Modeling
  • FIGS. 48 through 53 show various analyses of data performed using latent class modeling. From a targeted 104-gene panel, selected to be informative relative to biological state of MS patients (shown in Table 4), primers and probes were prepared for a subset of 54 genes (shown in Table 1B) (those with p-values of 0.05 or better) or 72 genes (shown in Tables 1B and 2 combined). Gene expression profiles were obtained using these subsets of genes, and of these individual genes, ITGAM was found to be uniquely and exquisitely informative regarding MS, yielding the best discrimination from normals of the genes examined.
  • In order, ranked by increasing p-values, with higher values indicating less discrimination from normals, the following genes shown in Table 1A were determined to be especially useful in discriminating MS subjects (all MS and 3-month washed out MS) from normals (listed below from more discriminating to less discriminating). A ranking of the top 54 genes is shown in Table 1B, listed from more discriminating to less discriminating, by p-value.
  • As shown above, ITGAM was shown to be most discriminating for MS, have the lowest p-value of all genes examined. Latent Class Modeling was then performed with several other genes in combination with ITGAM, to produce three-gene models, four-gene models, and 5-gene models for characterizing MS relative to normals data for a variety of MS subjects. These results are shown in FIGS. 48 through 53, discussed below.
  • FIG. 48 shows a three-gene model generated with Latent Class Modeling using ITGAM in combination with MMP9 and ITGA4. In this study, four different groups of MS subjects were compared to normals data for a subset of 72 genes of the 104-gene panel shown in Table 4. The question asked was, using only ITGAM combined with two other genes, in this case, MMP9 and ITGA4, is it possible to discriminate MS subjects from normal subjects (those with no history or diagnosis of MS) The groups of MS patients included “washed-out” subjects, i.e. those diagnosed with MS but off any treatment for three months or longer, and who are represented by Xs and diamonds. Another group of subjects, represented by pentagons, were MS subjects who were not washed out from treatment, but rather were on a treatment regimen at the time of this study. The subjects represented by circles were subjects from another clinical study diagnosed with MS and who were also on a treatment regimen at the time of this study. Within this group, two subjects “flared” during the study, and were put on different therapies, and thus moved towards the normal range, as indicated by data taken at that later time and represented in this figures as the star (mf10) and the flower (mf8). Normals data are represented by pentagons. As can be seen in the scatter plot depicted in FIG. 48, there is only moderate discrimination with this model between normals and MS subjects, although the discrimination between normals and “washed out” subjects is better.
  • FIG. 49 shows a scatter plot for an alternative three-gene model using ITGAM combined with CD4 and MMP9. The groups of MS patients included “washed out” subjects (Xs), subjects from one clinical study on a treatment regimen (triangle), subjects from another clinical study on a treatment regimen (squares), subjects on an experimental treatment regimen (diamonds), two subjects who flared during the study (mf8 and mf10), and normal subjects (circles). As can be seen, there is almost complete discrimination with this model between normals and “washed out” subjects. Less discrimination is observed, however, between normals and subjects from the other clinical studies who were being treated at the time these data were generated.
  • FIG. 50 shows a scatter plot of the same alternative three-gene model of FIG. 49 using ITGAM with MMP9 and CD4 but now displaying only washed out subjects relative to normals. As indicated by the straight line, there is almost complete discrimination with this model between normals (circles) and “washed out” (Xs) subjects.
  • FIG. 51 shows a scatter plot of a four-gene model useful for discriminating all MS subjects, whether washed out, on treatment, or pre-diagnosis. The four-gene model was produced using Latent Class Modeling with ITGAM with ITGA4, MMP9 and CALCA. As can be seen, most MS subjects analyzed (square, diamonds, circles) were quite well-discriminated from normals (pentagon) with this model.
  • FIG. 52 shows a scatter plot of a five-gene model using ITGAM with ITGA4, NFKB1B, MMP9 and CALCA which further discriminates all MS subjects (square diamonds, Xs) from normals (circles). Note that subjects designated as mf10 and mf8 can be seen to move closer to normal upon treatment during the study from their “flared” state which occurred after enrollment.
  • FIG. 53 shows a scatter plot of another five-gene model using ITGAM with ITGA4, NFKB1B, MMP9 and CXCR3 replacing CALCA. Because CALCA is a low expression gene in general, an alternative five-gene model was produced replacing CALCA with CXCR3. Again one can see how the two flared subjects, mf10 and mf8 move closer to normals (star and flower) after treatment. Normals (pentagon).
  • These data support illustrate that Gene Expression Profiles with sufficient precision and calibration as described herein (1) can determine subsets of individuals with a known biological condition, particularly individuals with multiple sclerosis or individuals with inflammatory conditions related to multiple sclerosis; (2) may be used to monitor the response of patients to therapy; (3) may be used to assess the efficacy and safety of therapy; and (4) may used to guide the medical management of a patient by adjusting therapy to bring one or more relevant Gene Expression Profiles closer to a target set of values, which may be normative values or other desired or achievable values. It has been shown that Gene Expression Profiles may provide meaningful information even when derived from ex vivo treatment of blood or other tissue. It has been shown that Gene Expression Profiles derived from peripheral whole blood are informative of a wide range of conditions neither directly nor typically associated with blood.
  • Gene Expression Profiles are used for characterization and monitoring of treatment efficacy of individuals with multiple sclerosis, or individuals with inflammatory conditions related to multiple sclerosis.
  • Additionally, Gene Expression Profiles are also used for characterization and early identification (including pre-symptomatic states) of infectious disease. This characterization includes discriminating between infected and uninfected individuals, bacterial and viral infections, specific subtypes of pathogenic agents, stages of the natural history of infection (e.g., early or late), and prognosis. Use of the algorithmic and statistical approaches discussed above to achieve such identification and to discriminate in such fashion is within the scope of various embodiments herein.
  • Example 13 Clinical Data Analyzed with Latent Class Modeling Together with Substantive Criteria
  • Using a targeted 104-gene panel, selected to be informative relative to biological state of MS patients (shown in Table 4), primers and probes were prepared for a subset of 24 genes identified in the Stepwise Regression Analysis shown in Table 3.
  • Gene expression profiles were obtained using these subsets of genes. Actual correct classification rate for the MS patients and the normal subjects was computed. Multi-gene models were constructed which were capable of correctly classifying MS and normal subjects with at least 75% accuracy. These results are shown in Tables 5-9 below. As demonstrated in Tables 6-9, a few as two genes allows discrimination between individuals with MS and normals at an accuracy of at least 75%.
  • One Gene Model
  • All 24 genes were evaluated for significance (i.e., p-value) regarding their ability to discriminate between MS and Normals, and ranked in the order of significance (see, Table 5). The optimal cutoff on the delta ct value for each gene was chosen that maximized the overall correct classification rate. The actual correct classification rate for the MS and Normal subjects was computed based on this cutoff and determined as to whether both reached the 75% criteria. None of these 1-gene models satisfied the 75%/75% criteria.
  • Two Gene Model
  • The top 8 genes (lowest p-value discriminating between MS and Normals) were subject to further analysis in a two-gene model. Each of the top 8 genes, one at a time, was used as the first gene in a 2-gene model, where all 23 remaining genes were evaluated as the second gene in this 2-gene model. (See Table 6). Column four illustrates the evaluated correct classification rates for these models (Data for those combinations of genes that fell below the 75%/75% cutoff, not all shown). The p-values in the 2-gene models assess the fit of the null hypothesis that the 2-gene model yields predictions of class memberships (MS vs. Normal) that are no different from chance predictions. The p-values were obtained from the SEARCH stepwise logistic procedure in the GOLDMineR program.
  • Also included in Table 6 is the R2 statistic provided by the GOLDMineR program, The R2 statistic is a less formal statistical measure of goodness of prediction, which varies between 0 (predicted probability of being in MS is constant regardless of delta-ct values on the 2 genes) to 1 (predicted probability of being MS=1 for each MS subject, and =0 for each Normal subject).
  • The right-most column of Table 6 indicates whether the 2-gene model was further used in illustrate the development of 3-gene models. For this use, 7 models with the lowest p-values (most significant), plus a few others were included as indicated.
  • Three Gene Model
  • For each of the selected 2-gene models (including the 7 most significant), each of the remaining 22 genes was evaluated as being included as a third gene in the model. Table 7 lists these along with the incremental p-value associated with the 3rd gene. Only models where the incremental p-value <0.05 are listed. The others were excluded because the additional MS vs. Normal discrimination associated with the 3rd gene was not significant at the 0.05 level. Each of these 3-gene models was evaluated further to determine whether incremental p-values associated with the other 2 genes was also significant. If the incremental p-value of any one of the 3 was found to be less than 0.05, it was excluded because it did not make a significant improvement over one of the 2-gene sub-models. An example of a 3-gene model that failed this secondary test was the model containing NFKB1B, HLADRA and CASP9. Here, the incremental p-value for NFKB1B was found to be only. 13 and therefore did not provide a significant improvement over the 2-gene model containing HLADRA and CASP9. The ESTIMATE procedure in GOLDMineR was used to compute all of the incremental p-values, which are shown in Table 7.
  • Four and Five Gene Models
  • The procedure for models containing 4 and five genes is similar to the one for three genes. Table 8 and 9 show the results associated with the use of most significant 3-gene model to obtain 4-gene and 5-gene models. The incremental p-values associated with each gene in the 4-gene and 5-gene models are shown, along with the percent classified correctly. As demonstrated by Tables 8 and 9 the addition of more genes in the model did not significantly alter the ability of the models to correctly classify MS patients and normals.
  • Example 14 Tests for Critical Unmet Needs in Rheumatology-Screening of Patients for MS Prior to anti-TNF Therapies
  • TNF inhibitors, including ENBREL, HUMERIA, REMICADE, and other agents that inhibit TNF, have been associated with rare cases of new or exacerbated symptoms of demyelinating disorders including but not limited to multiple sclerosis, and optic neuritis, seizure, neuromyelitis optica, transverse myelitis, acute disseminated encephalomyelitis, HIV encephalitis, adrenoleukodystrophy, adrenomyeloneuropathy, progress multifocal leukoencephalopathy, and central pontine myelinolysis, and CNS manifestations of systemic vasculitis, some case presenting with mental status changes and some associated with permanent disability. These TNF inhibitors have been shown to accelerate the demyelination process causing nerve lesions. For example, cases of transverse myelitis, optic neuritis, multiple sclerosis, and new onset or exacerbation of seizure disorders have been observed in association with ENBREL therapy. The causal relationship to ENBREL therapy remains unclear. While no clinical trials have been performed evaluating ENBREL therapy in patients with multiple sclerosis, other TNF antagonists administered to patients with multiple sclerosis have been associated with increases in disease activity. As such, prescribers should exercise caution in considering the use of ENBREL or other anti-TNF therapeutics in patients with preexisting or recent-onset central nervous system demyelinating disorders.
  • The present invention provides a method for predicting an adverse effect from anti-TNF therapy in a subject. The method comprises obtaining a sample from the subject (e.g., blood, tissue, or cell), the sample providing a source of RNAs, assessing a profile data set of a plurality of members, each member being a quantitative measure of the amount of a distinct RNA constituent in a panel of constituents (e.g., two or more constituents from any of Tables 1-10), selected so that measurement of the constituents enables characterization of the presumptive signs of a multiple sclerosis, wherein such measure for each constituent is obtained under measurement conditions that are substantially repeatable to produce a patient data set. This patient data set is then compared to a baseline profile data set, e.g. a profile data set multiple sclerosis or inflammatory conditions related to multiple sclerosis, determined as previous described. A patient data set that is similar to the baseline profile data set indicates the subject is at risk for suffering an adverse effect from anti-TNF therapy. The sample is obtained, prior to, during, or after administration of an anti-TNF therapeutic regimen.
  • In particular, the method is useful for screening subjects suffering from an inflammatory condition for a demyelinating disease prior to the administration of anti-TNF therapy for the treatment of the inflammatory condition. The inflammatory condition may include but are not limited to rheumatoid arthritis, psoriasis, ankylosing spondylitis, psoriatic arthritis and Crohn's disease. The demyelinating condition may include but is not limited to multiple sclerosis, optic neuritis, seizure, neuromyelitis optica, transverse myelitis, acute disseminated encephalomyelitis, HIV encephalitis, adrenoleukodystrophy, adrenomyeloneuropathy, progress multifocal leukoencephalopathy, and central pontine myelinolysis, and CNS manifestations of systemic vasculitis,
  • Examples of anti-TNF Therapeutics and Indications
  • Enbrel, containing etanercept, is a breakthrough product approved for the treatment of chronic inflammatory diseases such as rheumatoid arthritis, juvenile rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, and psoriasis. Enbrel continues to maintain a leading position in the dermatology and rheumatology biologic marketplaces, ranking No. 1 in worldwide sales among biotechnology products used in rheumatology and dermatology.
  • Abbott's Humira has been approved for treatment of rheumatoid arthritis in 57 countries, and for psoriatic arthritis and early RA in some European countries and the US.
  • Remicade has now achieved approvals in the treatment of such inflammatory diseases as Crohn's disease, rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis. First approved in 1998 for Crohn's disease, Remicade has been used to treat more than half a million patients worldwide.
  • RA Incidence and Prevalence Rates
  • Rheumatoid arthritis has a worldwide distribution with an estimated prevalence of 1 to 2%. Prevalence increases with age, approaching 5% in women over age 55. US prevalence of 2 million patients is 0.68%. The average annual incidence in the United States is about 70 per 100,000 annually. Over 200,000 new cases in US per year. Both incidence and prevalence of rheumatoid arthritis are two to three times greater in women than in men.
  • Psoriasis Incidence and Prevalence Rates
  • It is estimated that over seven million Americans (2.6%) have psoriasis, with more than 150,000 new cases reported each year.
  • Chronic plaque psoriasis represents approximately 80% of people with psoriasis with a US prevalence of approximately 5.7 million (2%). 10-20% of patients with plaque psoriasis also experience psoriatic arthritis.
  • TABLE 1A
    Normals vs. all MS sets Normals vs. 3-month washed out MS
    p-value p-value
    ITGAM 8.4E−21 ITGAM 2.7E−27
    NFKB1 1.1E−18 NFKB1 2.9E−18
    NFKBIB 1.4E−17 CASP9 3.8E−18
    CASP9 2.6E−15 IRF5 3.0E−17
    IRF5 3.0E−15 NFKBIB 2.1E−16
  • TABLE 1B
    Ranking of Genes, by P-Value, From More Discriminating to Less
    Discriminating
    p-value
    Gene p-value (Washed-
    # Symbol (MS v. N) out v. N)
    1 ITGAM 8.40E−21 2.70E−27
    2 NFKB1 1.10E−18 2.90E−18
    3 NFKBIB 1.40E−17 2.10E−16
    4 CASP9 2.60E−15 3.80E−18
    5 IRF5 3.00E−15 3.00E−17
    6 IL18R1 2.70E−12 1.50E−14
    7 TGFBR2 7.70E−12 1.30E−12
    8 NOS3 1.60E−10 1.50E−13
    9 IL1RN 2.00E−10 1.00E−07
    10 TLR2 5.70E−10 3.00E−08
    11 CXCR3 1.60E−09 2.00E−09
    12 FTL 2.00E−09 4.00E−09
    13 CCR1 3.60E−09 9.60E−07
    14 TNFSF13B 1.30E−08 2.90E−05
    15 TLR4 9.80E−08 2.10E−06
    16 LTA 2.20E−07 3.10E−10
    17 BCL2 2.50E−07 3.90E−08
    18 TREM1 6.20E−07 1.80E−05
    19 HMOX1 9.00E−07 2.40E−06
    20 CALCA 1.00E−06 8.00E−05
    21 PLAU 1.00E−06 4.30E−07
    22 TIMP1 1.10E−06 1.00E−06
    23 MIF 1.50E−06 1.30E−10
    24 PI3 8.40E−06 2.00E−09
    25 IL1B 5.50E−06 5.50E−06
    26 DTR 1.50E−05 0.00011
    27 CCL5 2.30E−05 6.90E−05
    28 IL13 4.60E−05 1.50E−06
    29 ARG2 5.10E−05 7.10E−06
    30 CCR5 5.80E−05 6.90E−05
    31 APAF1 7.60E−05 0.00016
    32 SERPINE1 8.30E−05 0.0001
    33 MMP3 9.90E−05 4.30E−5
    34 PLA2G7 0.00014 0.00043
    35 NOS1 0.00015 0.00041
    36 FCGR1A 0.00021 0.00041
    37 PF4 0.00032 2.70E−05
    38 ICAM1 0.00056 0.0016
    39 PTX3 0.00071 0.0014
    40 MMP9 0.00073 0.0012
    41 LBP 0.0011 6.60E−05
    42 MBL2 0.0014 0.00068
    43 CCL3 0.0039 0.011
    44 CXCL10 0.0043 1.00E−05
    45 PTGS2 0.0053 0.0025
    46 CD8A 0.0068 0.007
    47 SFTPD 0.0094 0.0089
    48 F3 0.015 0.0016
    49 CD4 0.018 0.0041
    50 CCL2 0.025 0.36
    51 IL6 0.027 0.05
    52 SPP1 0.029 0.012
    53 IL12B 0.03 0.011
    54 CASP1 0.045 0.26
  • TABLE 2
    Remaining Genes Making up the 72-gene Panel
    p-value
    Gene p-value (Washed-
    # Symbol (MS v. N) out v. N)
    55 TNFSF6 0.06 0.1
    56 ITGA4 0.08 0.23
    57 TNFSF5 0.085 0.23
    58 JUN 0.089 0.033
    59 CCR3 0.12 0.019
    60 CD86 0.12 0.62
    61 IFNG 0.15 0.2
    62 IL1A 0.15 0.057
    63 IL2 0.19 0.21
    64 IL8 0.21 0.3
    65 VEGF 0.39 0.2
    66 CASP3 0.41 0.5
    67 IL10 0.43 0.37
    68 CSF2 0.48 0.68
    69 CD19 0.56 0.94
    70 IL4 0.79 0.66
    71 CCL4 0.92 0.83
    72 IL15 0.94 0.81
  • TABLE 3
    Stepwise Regression Analysis of Wash-out MS Baseline Subjects
    (dataset A1A2, n = 103) vs Source MDx Normals (dataset N1, n = 100)
    LogIT p-value LogIT p-value LogIT p-value LogIT p-value
    Gene Loci (24) Step 1 Gene Loci (24) Step 2 Gene Loci (24) Step 3 Gene Loci (24) Step 4
    CASP9 3.20E−22 HLADRA 1.70E−10 ITGAL 8.60E−07 TGFBR2 5.20E−04
    ITGAM 2.40E−19 TGFBR2 1.70E−06 TGFBR2 9.10E−07 IL1R1 0.0025
    ITGAL 5.20E−18 ITGAL 0.0018 BCL2 0.0005 JUN 0.0084
    NFKBIB 1.20E−16 JUN 0.0024 IFI16 0.0065 ICAM1 0.043
    IL18R1 8.30E−16 VEGFB 0.0054 CD8A 0.0071 VEGFB 0.044
    NFKB1 8.60E−16 CD14 0.0066 IL18R1 0.013 IL18R1 0.048
    STAT3 7.60E−15 BCL2 0.0098 IL1R1 0.039 STAT3 0.048
    BCL2 4.00E−14 PI3 0.018 JUN 0.058 CD4 0.068
    IL1B 4.70E−11 IL18R1 0.02 PI3 0.16 CCR3 0.089
    PI3 6.20E−11 CCR3 0.059 MX1 0.16 PI3 0.11
    HSPA1A 5.80E−09 IL1R1 0.067 CD4 0.2 CD14 0.11
    CD4 1.30E−07 ICAM1 0.083 STAT3 0.21 HSPA1A 0.12
    ICAM1 3.40E−07 ITGAM 0.094 IL1B 0.29 IFI16 0.21
    TGFBR2 5.40E−07 IFI16 0.13 VEGFB 0.3 BCL2 0.28
    IFI16 5.60E−07 CD4 0.26 NFKBIB 0.3 NFKB1 0.31
    HLADRA 1.20E−05 CD8A 0.29 CCR3 0.32 CD8A 0.33
    IL1R1 5.70E−05 IL1B 0.42 BPI 0.53 ITGAM 0.47
    CD8A 6.30E−05 STAT3 0.5 HSPA1A 0.7 NFKBIB 0.59
    CD14 0.00018 HSPA1A 0.55 ICAM1 0.79 IL1B 0.77
    BPI 0.00085 NFKB1 0.9 CD14 0.98 MX1 0.83
    CCR3 0.0014 NFKBIB 0.91 ITGAM 0.99 BPI 0.94
    MX1 0.017 MX1 0.96 NFKB1 0.99 ITGAL included
    JUN 0.017 BPI 1 HLADRA included HLADRA included
    VEGFB 0.36 CASP9 included CASP9 included CASP9 included
    R-squared = 0.397 R-squared 0.544 R-squared 0.628 R-squared 0.669
    itgam + hladra R2 = 0.434
    itgal + hladra R2 = 0.55
    in this 3-gene model, hladra is most significant, itgal & casp9 are comparable
  • TABLE 4
    Precision Profile ™ for Multiple Sclerosis or Inflammatory Conditions Related
    to Multiple Sclerosis
    Symbol Name Classification Description
    APAF1 Apoptotic Protease Protease Cytochrome c binds to APAF1, triggering
    Activating Factor 1 activating activation of CASP3, leading to apoptosis.
    peptide May also facilitate procaspase 9 auto
    activation.
    ARG2 Arginase II Enzyme/redox Catalyzes the hydrolysis of arginine to
    ornithine and urea; may play a role in down
    regulation of nitric oxide synthesis
    BCL2 B-cell CLL/ Apoptosis Blocks apoptosis by interfering with the
    lymphoma 2 Inhibitor-cell activation of caspases
    cycle control-
    oncogenesis
    BPI Bactericidal/permeability- Membrane- LPS binding protein; cytotoxic for many gram
    increasing protein bound protease negative organisms; found in myeloid cells
    C1QA Complement Proteinase/ Serum complement system; forms C1
    component 1, q proteinase complex with the proenzymes c1r and c1s
    subcomponent, alpha inhibitor
    polypeptide
    CALCA Calcitonin/calcitonin- cell-signaling AKA CALC1; Promotes rapid incorporation
    related polypeptide, and activation of calcium into bone
    alpha
    CASP1 Caspase 1 Proteinase Activates IL1B; stimulates apoptosis
    CASP3 Caspase 3 Proteinase/ Involved in activation cascade of caspases
    Proteinase responsible for apoptosis - cleaves CASP6,
    Inhibitor CASP7, CASP9
    CASP9 Caspase 9 Proteinase Binds with APAF1 to become activated;
    cleaves and activates CASP3
    CCL1 Chemokine (C—C Cytokines- Secreted by activated T cells; chemotactic for
    Motif) ligand 1 chemokines- monocytes, but not neutrophils; binds to
    growth factors CCR8
    CCL2 Chemokine (C—C Cytokines- CCR2 chemokine; Recruits monocytes to
    Motif) ligand 2 chemokines- areas of injury and infection; Upregulated in
    growth factors liver inflammation; Stimulates IL-4
    production; Implicated in diseases involving
    monocyte, basophil infiltration of tissue (e.g.
    psoriasis, rheumatoid arthritis,
    atherosclerosis)
    CCL3 Chemokine (C—C Cytokines- AKA: MIP1-alpha; monokine that binds to
    motif) ligand 3 chemokines- CCR1, CCR4 and CCR5; major HIV-
    growth factors suppressive factor produced by CD8 cells.
    CCL4 Chemokine (C—C Cytokines- Inflammatory and chemotactic monokine;
    Motif) ligand 4 chemokines- binds to CCR5 and CCR8
    growth factors
    CCL5 Chemokine (C—C Cytokines- Binds to CCR1, CCR3, and CCR5 and is a
    Motif) ligand 5 chemokines- chemoattractant for blood monocytes,
    growth factors memory T-helper cells and eosinophils; A
    major HIV-suppressive factor produced by
    CD8-positive T-cells
    CCR1 chemokine (C—C chemokine A member of the beta chemokine receptor
    motif) receptor 1 receptor family (seven transmembrane protein). Binds
    SCYA3/MIP-1a, SCYA5/RANTES, MCP-3,
    HCC-1, 2, and 4, and MPIF-1. Plays role in
    dendritic cell migration to inflammation sites
    and recruitment of monocytes.
    CCR3 Chemokine (C—C Chemokine C—C type chemokine receptor (Eotaxin
    motif) receptor 3 receptor receptor) binds to Eotaxin, Eotaxin-3, MCP-3,
    MCP-4, SCYA5/RANTES and mip-1 delta
    thereby mediating intracellular calcium flux.
    Alternative co-receptor with CD4 for HIV-1
    infection. Involved in recruitment of
    eosinophils. Primarily a Th2 cell chemokine
    receptor.
    CCR5 chemokine (C—C chemokine Binds to CCL3/MIP-1a and CCL5/RANTES.
    motif) receptor 5 receptor An important co-receptor for macrophage-
    tropic virus, including HIV, to enter cells.
    CD14 CD14 antigen Cell Marker LPS receptor used as marker for monocytes
    CD19 CD19 antigen Cell Marker AKA Leu 12; B cell growth factor
    CD3Z CD3 antigen, zeta Cell Marker T-cell surface glycoprotein
    polypeptide
    CD4 CD4 antigen (p55) Cell Marker Helper T-cell marker
    CD86 CD
    86 Antigen (cD Cell signaling AKA B7-2; membrane protein found in B
    28 antigen ligand) and activation lymphocytes and monocytes; co-stimulatory
    signal necessary for T lymphocyte
    proliferation through IL2 production.
    CD8A CD8 antigen, alpha Cell Marker Suppressor T cell marker
    polypeptide
    CKS2 CDC28 protein kinase Cell signaling Essential for function of cyclin-dependent
    regulatory subunit 2 and activation kinases
    CRP C-reactive protein acute phase the function of CRP relates to its ability to
    protein recognize specifically foreign pathogens and
    damaged cells of the host and to initiate their
    elimination by interacting with humoral and
    cellular effector systems in the blood
    CSF2 Granulocyte- Cytokines- AKA GM-CSF; Hematopoietic growth factor;
    monocyte colony chemokines- stimulates growth and differentiation of
    stimulating factor growth factors hematopoietic precursor cells from various
    lineages, including granulocytes,
    macrophages, eosinophils, and erythrocytes
    CSF3 Colony stimulating Cytokines- AKA GCSF controls production
    factor 3 (granulocyte) chemokines- differentiation and function of granulocytes.
    growth factors
    CXCL3 Chemokine Cytokines- Chemotactic pro-inflammatory activation-
    (C—X—C-motif) ligand 3 chemokines- inducible cytokine, acting primarily upon
    growth factors hemopoietic cells in immunoregulatory
    processes, may also play a role in
    inflammation and exert its effects on
    endothelial cells in an autocrine fashion.
    CXCL10 Chemokine (C—X—C Cytokines- AKA: Gamma IP10; interferon inducible
    motif) ligand 10 chemokines- cytokine IP10; SCYB10; Ligand for CXCR3;
    growth factors binding causes stimulation of monocytes, NK
    cells; induces T cell migration
    CXCR3 chemokine (C—X—C cytokines- Binds to SCYB10/IP-10, SCYB9/MIG,
    motif) receptor 3 chemokines- SCYB11/I-TAC. Binding of chemokines to
    growth factors CXCR3 results in integrin activation,
    cytoskeletal changes and chemotactic
    migration.
    DPP4 Dipeptidyl-peptidase 4 Membrane Removes dipeptides from unmodified, n-
    protein; terminus prolines; has role in T cell activation
    exopeptidase
    DTR Diphtheria toxin cell signaling, Thought to be involved in macrophage-
    receptor (heparin- mitogen mediated cellular proliferation. DTR is a
    binding epidermal potent mitogen and chemotactic factor for
    growth factor-like fibroblasts and smooth muscle cells, but not
    growth factor) endothelial cells.
    ELA2 Elastase 2, neutrophil Protease Modifies the functions of NK cells,
    monocytes and granulocytes
    F3 F3 enzyme/redox AKA thromboplastin, Coagulation Factor 3;
    cell surface glycoprotein responsible for
    coagulation catalysis
    FCGR1A Fc fragment of IgG, Membrane Membrane receptor for CD64; found in
    high affinity receptor protein monocytes, macrophages and neutrophils
    IA
    FTL Ferritin, light iron chelator Intracellular, iron storage protein
    polypeptide
    GZMB Granzyme B proteinase AKA CTLA1; Necessary for target cell lysis
    in cell-mediated immune responses. Crucial
    for the rapid induction of target cell apoptosis
    by cytotoxic T cells. Inhibition of the
    GZMB-IGF2R (receptor for GZMB)
    interaction prevented GZMB cell surface
    binding, uptake, and the induction of
    apoptosis.
    HLA-DRA Major Membrane Anchored heterodimeric molecule; cell-
    Histocompatability protein surface antigen presenting complex
    Complex; class II, DR
    alpha
    HMOX1 Heme oxygenase Enzyme/ Endotoxin inducible
    (decycling) 1 Redox
    HSPA1A Heat shock protein 70 Cell Signaling heat shock protein 70 kDa; Molecular
    and activation chaperone, stabilizes AU rich mRNA
    HIST1H1C Histo
    1, Hic Basic nuclear responsible for the nucleosome structure
    protein within the chromosomal fiber in eukaryotes;
    may attribute to modification of nitrotyrosine-
    containing proteins and their
    immunoreactivity to antibodies against
    nitrotyrosine.
    ICAM1 Intercellular adhesion Cell Adhesion/ Endothelial cell surface molecule; regulates
    molecule 1 Matrix Protein cell adhesion and trafficking, unregulated
    during cytokine stimulation
    IFI16 Gamma interferon Cell signaling Transcriptional repressor
    inducible protein
    16 and activation
    IFNA2 Interferon, alpha 2 Cytokines- interferon produced by macrophages with
    chemokines- antiviral effects
    growth factors
    IFNG Interferon, Gamma Cytokines/ Pro- and anti-inflammatory activity; TH1
    Chemokines/ cytokine; nonspecific inflammatory mediator;
    Growth Factors produced by activated T-cells.
    IL10 Interleukin 10 Cytokines- Anti-inflammatory; TH2; suppresses
    chemokines- production of proinflammatory cytokines
    growth factors
    IL12B Interleukin
    12 p40 Cytokines- Proinflammatory; mediator of innate
    chemokines- immunity, TH1 cytokine, requires co-
    growth factors stimulation with IL-18 to induce IFN-g
    IL13 Interleukin
    13 Cytokines/ Inhibits inflammatory cytokine production
    Chemokines/
    Growth Factors
    IL18 Interleukin
    18 Cytokines- Proinflammatory, TH1, innate and acquired
    chemokines- immunity, promotes apoptosis, requires co-
    growth factors stimulation with IL-1 or IL-2 to induce TH1
    cytokines in T- and NK-cells
    IL18R1 Interleukin
    18 Membrane Receptor for interleukin 18; binding the
    receptor 1 protein agonist leads to activation of NFKB-B;
    belongs to IL1 family but does not bind IL1A
    or IL1B.
    IL1A Interleukin 1, alpha Cytokines- Proinflammatory; constitutively and inducibly
    chemokines- expressed in variety of cells. Generally
    growth factors cytosolic and released only during severe
    inflammatory disease
    IL1B Interleukin
    1, beta Cytokines- Proinflammatory; constitutively and inducibly
    chemokines- expressed by many cell types, secreted
    growth factors
    IL1R1 Interleukin
    1 receptor, Cell signaling AKA: CD12 or IL1R1RA; Binds all three
    type I and activation forms of interleukin-1 (IL1A, IL1B and
    IL1RA). Binding of agonist leads to NFKB
    activation
    IL1RN Interleukin 1 Cytokines/ IL1 receptor antagonist; Anti-inflammatory;
    Receptor Antagonist Chemokines/ inhibits binding of IL-1 to IL-1 receptor by
    Growth Factors binding to receptor without stimulating IL-1-
    like activity
    IL2 Interleukin 2 Cytokines/ T-cell growth factor, expressed by activated
    Chemokines/ T-cells, regulates lymphocyte activation and
    Growth Factors differentiation; inhibits apoptosis, TH1
    cytokine
    IL4 Interleukin 4 Cytokines/ Anti-inflammatory; TH2; suppresses
    Chemokines/ proinflammatory cytokines, increases
    Growth Factors expression of IL-1RN, regulates lymphocyte
    activation
    IL5 Interleukin 5 Cytokines/ Eosinophil stimulatory factor; stimulates late
    Chemokines/ B cell differentiation to secretion of Ig
    Growth Factors
    IL6 Interleukin 6 Cytokines- Pro- and anti-inflammatory activity, TH2
    (interferon, beta 2) chemokines- cytokine, regulates hematopoietic system and
    growth factors activation of innate response
    IL8 Interleukin 8 Cytokines- Proinflammatory, major secondary
    chemokines- inflammatory mediator, cell adhesion, signal
    growth factors transduction, cell-cell signaling, angiogenesis,
    synthesized by a wide variety of cell types
    IL15 Interleukin 15 cytokines- Proinflammatory, mediates T-cell activation,
    chemokines- inhibits apoptosis, synergizes with IL-2 to
    growth factors induce IFN-g and TNF-a
    IRF5 interferon regulatory Transcription possess a novel helix-turn-helix DNA-binding
    factor 5 factor motif and mediate virus- and interferon
    (IFN)-induced signaling pathways.
    IRF7 Interferon regulatory Transcription Regulates transcription of interferon genes
    factor 7 Factor through DNA sequence-specific binding.
    Diverse roles include virus-mediated
    activation of interferon, and modulation of
    cell growth, differentiation, apoptosis, and
    immune system activity.
    ITGA-4 integrin alpha 4 integrin receptor for fibronectin and VCAM1; triggers
    homotypic aggregation for VLA4 positive
    leukocytes; participates in cytolytic T-cell
    interactions with target cells.
    ITGAM Integrin, alpha M; integrin AKA: Complement receptor, type 3, alpha
    complement receptor subunit; neutrophil adherence receptor; role in
    adherence of neutrophils and monocytes to
    activate endothelium
    LBP Lipopolysaccharide membrane Acute phase protein; membrane protein that
    binding protein protein binds to Lipid a moiety of bacterial LPS
    LTA LTA (lymphotoxin Cytokine Cytokine secreted by lymphocytes and
    alpha) cytotoxic for a range of tumor cells; active in
    vitro and in vivo
    LTB Lymphotoxin beta Cytokine Inducer of inflammatory response and normal
    (TNFSF3) lymphoid tissue development
    JUN v-jun avian sarcoma Transcription Proto-oncoprotein; component of
    virus 17 oncogene factor-DNA transcription factor AP-1 that interacts
    homolog binding directly with target DNA sequences to
    regulate gene expression
    MBL2 Mannose-binding lectin AKA: MBP1; mannose binding protein C
    protein precursor
    MIF Macrophage Cell signaling AKA; GIF; lymphokine, regulators
    migration inhibitory and growth macrophage functions through suppression of
    factor factor anti-inflammatory effects of glucocorticoids
    MMP9 Matrix proteinase AKA gelatinase B; degrades extracellular
    metalloproteinase
    9 matrix molecules, secreted by IL-8-stimulated
    neutrophils
    MMP3 Matrix proteinase capable of degrading proteoglycan,
    metalloproteinase 3 fibronectin, laminin, and type IV collagen,
    but not interstitial type I collagen.
    MX1 Myxovirus resistance peptide Cytoplasmic protein induced by influenza;
    1; interferon inducible associated with MS
    protein p78
    N33 Putative prostate Tumor Integral membrane protein. Associated with
    cancer tumor Suppressor homozygous deletion in metastatic prostate
    suppressor cancer.
    NFKB1 Nuclear factor of Transcription p105 is the precursor of the p50 subunit of the
    kappa light Factor nuclear factor NFKB, which binds to the
    polypeptide gene kappa-b consensus sequence located in the
    enhancer in B-cells 1 enhancer region of genes involved in immune
    (p105) response and acute phase reactions; the
    precursor does not bind DNA itself
    NFKBIB Nuclear factor of Transcription Inhibits/regulates NFKB complex activity by
    kappa light Regulator trapping NFKB in the cytoplasm.
    polypeptide gene Phosphorylated serine residues mark the
    enhancer in B-cells NFKBIB protein for destruction thereby
    inhibitor, beta allowing activation of the NFKB complex.
    NOS1 nitric oxide synthase enzyme/redox synthesizes nitric oxide from L-arginine and
    1 (neuronal) molecular oxygen, regulates skeletal muscle
    vasoconstriction, body fluid homeostasis,
    neuroendocrine physiology, smooth muscle
    motility, and sexual function
    NOS3 Nitric oxide synthase 3 enzyme/redox enzyme found in endothelial cells mediating
    smooth muscle relation; promotes clotting
    through the activation of platelets
    PAFAH1B1 Platelet activating Enzyme Inactivates platelet activating factor by
    factor removing the acetyl group
    acetylhydrolase,
    isoform !b, alpha
    subunit; 45 kDa
    PF4 Platelet Factor 4 Chemokine PF4 is released during platelet aggregation
    (SCYB4) and is chemotactic for neutrophils and
    monocytes. PF4's major physiologic role
    appears to be neutralization of heparin-like
    molecules on the endothelial surface of blood
    vessels, thereby inhibiting local antithrombin
    III activity and promoting coagulation.
    PI3 Proteinase inhibitor 3 Proteinase aka SKALP; Proteinase inhibitor found in
    skin derived inhibitor- epidermis of several inflammatory skin
    protein binding- diseases; it's expression can be used as a
    extracellular marker of skin irritancy
    matrix
    PLA2G7 Phospholipase A2, Enzyme/ Platelet activating factor
    group VII (platelet Redox
    activating factor
    acetylhydrolase,
    plasma)
    PLAU Plasminogen proteinase AKA uPA; cleaves plasminogen to plasmin (a
    activator, urokinase protease responsible for nonspecific
    extracellular matrix degradation; UPA
    stimulates cell migration via a UPA receptor
    PLAUR plasminogen Membrane key molecule in the regulation of cell-surface
    activator, urokinase protein; plasminogen activation; also involved in cell
    receptor receptor signaling.
    PTGS2 Prostaglandin- Enzyme Key enzyme in prostaglandin biosynthesis
    endoperoxide and induction of inflammation
    synthase 2
    PTX3 Pentaxin-related gene, Acute Phase AKA TSG-14; Pentaxin 3; Similar to the
    rapidly induced by Protein pentaxin subclass of inflammatory acute-
    IL-1 beta phase proteins; novel marker of inflammatory
    reactions
    RAD52 RAD52 (S. cerevisiae) DNA binding Involved in DNA double-stranded break
    homolog proteins or repair and meiotic/mitotic recombination
    SERPINE1 Serine (or cysteine) Proteinase/ Plasminogen activator inhibitor-1/PAI-1
    protease inhibitor, Proteinase
    class B (ovalbumin), Inhibitor
    member 1
    SFTPD Surfactant, pulmonary extracellular AKA: PSPD; mannose-binding protein;
    associated protein D lipoprotein suggested role in innate immunity and
    surfactant metabolism
    SLC7A1 Solute carrier family Membrane High affinity, low capacity permease involved
    7, member 1 protein; in the transport of positively charged amino
    permease acids
    SPP1 secreted cell signaling binds vitronectin; protein ligand of CD44,
    phosphoprotein 1 and activation cytokine for type 1 responses mediated by
    (osteopontin) macrophages
    STAT3 Signal transduction Transcription AKA APRF: Transcription factor for acute
    and activator of factor phase response genes; rapidly activated in
    transcription 3 response to certain cytokines and growth
    factors; binds to IL6 response elements
    TGFBR2 Transforming growth Membrane AKA: TGFR2; membrane protein involved in
    factor, beta receptor II protein cell signaling and activation, ser/thr protease;
    binds to DAXX.
    TIMP1 Tissue inhibitor of Proteinase/ Irreversibly binds and inhibits
    metalloproteinase 1 Proteinase metalloproteinases, such as collagenase
    Inhibitor
    TLR2 toll-like receptor 2 cell signaling mediator of peptidoglycan and lipotechoic
    and activation acid induced signaling
    TLR4 Toll-like receptor 4 Cell signaling mediator of LPS induced signaling
    and activation
    TNF Tumor necrosis factor Cytokine/tumor Negative regulation of insulin action.
    necrosis factor Produced in excess by adipose tissue of obese
    receptor ligand individuals - increases IRS-1 phosphorylation
    and decreases insulin receptor kinase activity.
    Pro-inflammatory; TH1 cytokine; Mediates
    host response to bacterial stimulus; Regulates
    cell growth & differentiation
    TNFRSF7 Tumor necrosis factor Membrane Receptor for CD27L; may play a role in
    receptor superfamily, protein; activation of T cells
    member
    7 receptor
    TNFSF13B Tumor necrosis factor Cytokines- B cell activating factor, TNF family
    (ligand) superfamily, chemokines-
    member 13b growth factors
    TNFRSF13B Tumor necrosis factor Cytokines- B cell activating factor, TNF family
    receptor superfamily, chemokines-
    member 13, subunit growth factors
    beta
    TNFSF5 Tumor necrosis factor Cytokines- Ligand for CD40; expressed on the surface of
    (ligand) superfamily, chemokines- T cells. It regulates B cell function by
    member 5 growth factors engaging CD40 on the B cell surface.
    TNFSF6 Tumor necrosis factor Cytokines- AKA FasL; Ligand for FAS antigen;
    (ligand) superfamily, chemokines- transduces apoptotic signals into cells
    member
    6 growth factors
    TREM1 Triggering receptor cell signaling Member of the Ig superfamily; receptor
    expressed on myeloid and activation exclusively expressed on myeloid cells.
    cells 1 TREM1 mediates activation of neutrophils
    and monocytes and may have a predominant
    role in inflammatory responses
    VEGF vascular endothelial cytokines- VPF; Induces vascular permeability,
    growth factor chemokines- endothelial cell proliferation, angiogenesis.
    growth factors Produced by monocytes
  • TABLE 5
    Ranking of select genes from Table 4 (from most to least significant),
    based on 1-WAY ANOVA approach
    gene p-value
    CASP9 1.80E−19
    ITGAL 3.00E−19
    ITGAM 3.40E−16
    STAT3 2.10E−15
    NFKB1 2.90E−15
    NFKBIB 5.60E−14
    HLADRA 1.00E−11
    BCL2 5.40E−11
    IL1B 2.30E−10
    PI3 3.10E−10
    IFI16 3.30E−10
    IL18R1 7.80E−10
    HSPA1A 2.00E−08
    ICAM1 1.90E−07
    TGFBR2 4.80E−06
    CD4 3.30E−05
    BPI 6.20E−05
    IL1R1 0.0001
    CD14 0.00082
    CD8A 0.0012
    MX1 0.0076
    JUN 0.027
    CCR3 0.13
    VEGFB 0.58
  • TABLE 6
    2 gene models capable of correctly classifying MS v. Normal Subjects
    Correct used to
    Classification illustrate
    % 3-gene
    gene1 gene2 p-value % MS normals R2 models?
    ITGAL HLADRA 1.6E−39 85.4% 82.9% 0.531 YES
    CASP9 HLADRA 1.9E−35 78.5% 84.2% 0.478 YES
    NFKBIB HLADRA 1.9E−31 80.0% 80.9% 0.429 YES
    STAT3 HLADRA 2.9E−31 77.7% 86.2% 0.428 YES
    NFKB1 HLADRA 3.0E−29 82.3% 80.3% 0.401 YES
    ITGAM HLADRA 1.6E−28 80.0% 80.9% 0.405 YES
    ITGAL VEGFB 7.3E−28 77.7% 80.9% 0.383 YES
    HLADRA BCL2 5.3E−27 76.2% 82.9% 0.374
    HLADRA CD4 8.3E−26 83.1% 75.0% 0.357
    HLADRA IL1B 1.1E−24 74.6% 79.6% 0.342
    HLADRA HSPA1A 1.3E−24 76.9% 77.6% 0.340
    HLADRA ICAM1 9.9E−24 76.2% 77.0% 0.331
    CASP9 VEGFB 1.4E−22 75.4% 77.0% 0.317
    HLADRA IL18R1 1.4E−22 76.2% 79.6% 0.316
    CASP9 TGFBR2 5.0E−22 75.4% 73.7% 0.319 YES
    HLADRA CD14 1.9E−21 75.4% 73.7% 0.300
    CASP9 ITGAL 2.0E−21 73.8% 70.4% 0.303
    ITGAL PI3 2.8E−21 80.0% 75.7% 0.302
    HLADRA IFI16 3.4E−21 75.4% 75.0% 0.296
    CASP9 CCR3 3.9E−21 72.3% 75.0% 0.296
    ITGAL CD4 7.8E−21 76.2% 71.1% 0.293
    CASP9 IFI16 8.4E−21 75.4% 74.3% 0.292 YES
    ITGAL ITGAM 1.4E−20 76.2% 75.7% 0.303
    STAT3 CD14 2.1E−20 74.6% 75.0% 0.286
    CASP9 CD14 2.6E−20 74.6% 75.7% 0.286
    CASP9 PI3 2.7E−20 70.8% 77.0% 0.287
    ITGAL CD14 4.6E−20 76.2% 71.7% 0.284
    ITGAL IFI16 5.5E−20 77.7% 71.1% 0.283
    ITGAL CCR3 9.6E−20 0.280
    CASP9 JUN 1.2E−19 76.2% 76.3% 0.290
    BCL2 VEGFB 1.8E−19 76.2% 73.0% 0.274
    CASP9 CD4 2.1E−19 74.6% 67.1% 0.274
    ITGAL NFKB1 2.2E−19 75.4% 71.7% 0.276
    ITGAL IL1B 2.9E−19 75.4% 72.4% 0.273
    ITGAL NFKBIB 3.9E−19 70.8% 75.7% 0.273
    CASP9 BCL2 4.7E−19 72.3% 73.0% 0.270
    ITGAL JUN 4.7E−19 0.281
    ITGAL IL18R1 6.6E−19 75.4% 69.1% 0.269
    CASP9 STAT3 6.7E−19 76.2% 71.7% 0.267
    CASP9 IL1R1 7.9E−19 72.3% 73.7% 0.266
    HLADRA PI3 1.0E−18 74.6% 73.0% 0.261
    CASP9 IL1B 1.1E−18 77.7% 69.1% 0.265
    ITGAL STAT3 1.1E−18 70.0% 74.3% 0.266
    ITGAL CD8A 1.1E−18 70.0% 76.3% 0.266
    ITGAM IFI16 1.3E−18 75.4% 76.3% 0.275
    CASP9 ICAM1 1.4E−18 74.6% 74.3% 0.263
    CASP9 BPI 1.4E−18 76.2% 71.1% 0.264
    NFKB1 VEGFB 1.5E−18 76.9% 69.1% 0.263
    CASP9 CD8A 1.7E−18 73.8% 74.3% 0.262
    CASP9 NFKB1 1.8E−18 75.4% 72.4% 0.262
    ITGAL BCL2 1.8E−18 0.264
    CASP9 NFKBIB 1.9E−18 77.7% 69.7% 0.261
    CASP9 IL18R1 2.0E−18 70.8% 75.0% 0.261
    CASP9 HSPA1A 2.0E−18 72.3% 73.7% 0.261
    ITGAL ICAM1 2.2E−18 73.1% 71.7% 0.262
    ITGAL BPI 2.2E−18 72.3% 73.7% 0.262
    ITGAL IL1R1 2.7E−18 70.8% 77.0% 0.261
    HLADRA TGFBR2 2.8E−18 74.6% 75.0% 0.269
    CASP9 ITGAM 2.9E−18 75.4% 73.0% 0.271
    ITGAL HSPA1A 3.4E−18 75.4% 69.7% 0.260
    ITGAL TGFBR2 3.8E−18 75.4% 71.7% 0.270
    CASP9 MX1 4.0E−18 75.4% 71.1% 0.268
    ITGAL MX1 9.0E−18 73.8% 73.0% 0.265
    HLADRA CD8A 1.1E−17 74.6% 67.1% 0.248
    ITGAM BCL2 5.2E−17 69.2% 78.9% 0.254
    ITGAM CD14 3.5E−16 68.5% 76.3% 0.243
    ITGAM TGFBR2 5.5E−16 75.4% 76.3% 0.240
    NFKBIB TGFBR2 9.6E−14 73.8% 74.3% 0.222
  • TABLE 7
    3 gene models capable of correctly classifying MS v. Normal Subjects
    incremental incremental incremental
    p-value p-value p-value
    gene p-value p-value R-squared % MS % normals gene p-value gene p-value
    ITGAL HLADRA CASP9 0.00024 2.10E−41 0.563 85.4% 86.8%
    ITGAL HLADRA NFKBIB 0.003 2.20E−40 0.553 81.5% 88.2%
    ITGAL HLADRA IL1B 0.0061 4.10E−40 0.549 85.4% 84.9%
    ITGAL HLADRA ITGAM 0.02 2.20E−39 0.552 86.2% 84.9%
    ITGAL HLADRA VEGFB 0.021 1.20E−39 0.544 83.1% 86.2%
    ITGAL HLADRA PI3 0.03 1.70E−39 0.543 83.8% 84.9%
    CASP9 HLADRA ITGAL 1.40E−08 2.10E−41 0.563 85.4% 86.8%
    CASP9 HLADRA TGFBR2 0.00048 2.60E−36 0.515 83.8% 82.2%
    CASP9 HLADRA BCL2 0.00056 5.20E−37 0.509 85.4% 81.6%
    CASP9 HLADRA IFI16 0.0016 1.30E−36 0.506 83.1% 84.9%
    CASP9 HLADRA CD8A 0.0043 3.30E−36 0.499 83.8% 80.9%
    CASP9 HLADRA STAT3 0.022 1.40E−35 0.493 82.3% 82.2%
    CASP9 HLADRA CCR3 0.03 1.80E−35 0.489 81.5% 80.9%
    CASP9 HLADRA MX1 0.034 4.40E−35 0.497 83.1% 80.3%
    NFKBIB HLADRA ITGAL 1.20E−11 2.20E−40 0.553 81.5% 88.2%
    NFKBIB HLADRA BCL2 1.10E−06 1.40E−35 0.492 80.0% 83.6%
    NFKBIB HLADRA STAT3 5.20E−06 6.10E−35 0.484 80.8% 81.6%
    NFKBIB HLADRA CASP9 5.40E−06 6.30E−35 0.483 77.7% 81.6% nfkbib 0.13 hladra 2.80E−19
    NFKBIB HLADRA IL1B 0.00028 2.60E−33 0.464 79.2% 84.2%
    NFKBIB HLADRA IFI16 0.00039 3.50E−33 0.464 77.7% 84.9%
    NFKBIB HLADRA HSPA1A 0.0004 3.60E−33 0.461 79.2% 80.9% nfkbib 3.40E−11
    NFKBIB HLADRA CD4 0.00043 3.90E−33 0.462 79.2% 80.9%
    NFKBIB HLADRA BPI 0.0043 3.20E−32 0.449 79.2% 82.9% nfkbib 3.70E−18
    NFKBIB HLADRA MX1 0.0045 5.80E−32 0.458 80.0% 83.6% nfkbib 2.20E−20
    NFKBIB HLADRA IL18R1 0.0046 3.40E−32 0.45 77.7% 82.9%
    NFKBIB HLADRA ITGAM 0.0053 2.10E−31 0.45 80.0% 82.9%
    NFKBIB HLADRA CD8A 0.0068 4.80E−32 0.449 78.5% 83.6% nfkbib 4.10E−17
    NFKBIB HLADRA ICAM1 0.015 9.70E−32 0.445 77.7% 81.6%
    NFKBIB HLADRA TGFBR2 0.019 6.20E−31 0.445 77.7% 81.6% nfkbib 2.20E−15
    NFKBIB HLADRA NFKB1 0.021 1.30E−31 0.443 77.7% 83.6% nfkbib 8.40E−05
    NFKBIB HLADRA CD14 0.036 2.10E−31 0.441 77.7% 82.2%
    NFKBIB HLADRA PI3 0.049 2.70E−31 0.438 76.9% 83.6%
    STAT3 HLADRA ITGAL 2.70E−10 6.70E−39 0.535 82.3% 86.2%
    STAT3 HLADRA BCL2 4.30E−07 8.40E−36 0.495 83.1% 87.5% STAT3 1.80E−11
    STAT3 HLADRA CASP9 7.40E−07 1.40E−35 0.493 80.0% 84.2%
    STAT3 HLADRA NFKBIB 3.40E−06 6.10E−35 0.484 79.2% 83.6% STAT3 5.20E−06
    STAT3 HLADRA IL1R1 1.80E−05 3.00E−34 0.473 79.2% 81.6% STAT3 4.00E−21
    STAT3 HLADRA CD8A 0.00012 1.80E−33 0.466 79.2% 80.3% STAT3 1.40E−18
    STAT3 HLADRA NFKB1 0.00057 7.60E−33 0.46 80.8% 84.2% STAT3 4.30E−06
    STAT3 HLADRA ITGAM 0.0062 4.10E−31 0.45 81.5% 84.2%
    STAT3 HLADRA IFI16 0.0062 6.70E−32 0.449 80.0% 83.6%
    STAT3 HLADRA CD4 0.0097 1.00E−31 0.446 81.5% 83.6%
    STAT3 HLADRA PI3 0.012 1.20E−31 0.445 80.0% 82.9%
    STAT3 HLADRA IL18R1 0.021 2.00E−31 0.442 80.8% 84.2%
    NFKB1 HLADRA ITGAL 2.00E−12 5.90E−39 0.537 83.8% 86.2%
    NFKB1 HLADRA CASP9 7.90E−08 1.70E−34 0.479 79.2% 84.2%
    NFKB1 HLADRA STAT3 4.30E−06 7.60E−33 0.46 80.8% 84.2%
    NFKB1 HLADRA NFKBIB 8.40E−05 1.30E−31 0.443 77.7% 83.6% NFKB1 0.021
    NFKB1 HLADRA BCL2 0.00022 3.20E−31 0.439 76.9% 82.9% NFKB1 9.80E−07
    NFKB1 HLADRA HSPA1A 0.00042 5.70E−31 0.435 78.5% 82.9% NFKB1 6.20E−09
    NFKB1 HLADRA IL1B 0.00051 6.80E−31 0.435 78.5% 81.6%
    NFKB1 HLADRA IFI16 0.0009 1.20E−30 0.43 81.5% 85.5%
    NFKB1 HLADRA ITGAM 0.0018 1.10E−29 0.43 78.5% 82.9%
    NFKB1 HLADRA ICAM1 0.0028 3.30E−30 0.426 78.5% 82.9%
    NFKB1 HLADRA CD8A 0.0049 5.40E−30 0.424 77.7% 83.6% NFKB1 5.10E−15
    NFKB1 HLADRA BPI 0.0079 8.30E−30 0.419 77.7% 84.9% NFKB1 1.10E−15
    NFKB1 HLADRA CD4 0.011 1.10E−29 0.419 78.5% 83.6%
    NFKB1 HLADRA MX1 0.016 2.50E−29 0.425 80.0% 82.9% NFKB1 1.10E−17
    NFKB1 HLADRA PI3 0.018 1.70E−29 0.416 78.5% 84.2%
    NFKB1 HLADRA IL18R1 0.025 2.30E−29 0.415 79.2% 80.3%
    ITGAM HLADRA ITGAL 1.40E−13 2.20E−39 0.552 86.2% 84.9%
    ITGAM HLADRA CASP9 8.90E−08 8.80E−34 0.481 78.5% 82.2%
    ITGAM HLADRA BCL2 2.50E−07 2.40E−33 0.476 78.5% 83.6% ITGAM 1.00E−09
    ITGAM HLADRA IFI16 1.70E−05 1.30E−31 0.456 82.3% 82.9%
    ITGAM HLADRA NFKBIB 2.80E−05 2.10E−31 0.45 80.0% 82.9% ITGAM 0.0053
    ITGAM HLADRA STAT3 5.80E−05 4.10E−31 0.45 81.5% 84.2%
    ITGAM HLADRA CD8A 0.00028 1.80E−30 0.441 80.0% 82.9% ITGAM 3.60E−16
    ITGAM HLADRA CD4 0.00078 4.50E−30 0.437 79.2% 83.6%
    ITGAM HLADRA NFKB1 0.0021 1.10E−29 0.43 78.5% 82.9%
    ITGAM HLADRA IL1B 0.0046 2.30E−29 0.427 77.7% 82.2%
    ITGAM HLADRA MX1 0.0054 2.90E−29 0.435 80.8% 83.6% ITGAM 2.90E−18
    ITGAM HLADRA PI3 0.031 1.20E−28 0.417 77.7% 82.2%
    ITGAM HLADRA VEGFB 0.031 1.20E−28 0.417 78.5% 83.6% ITGAM 5.60E−18
    ITGAM HLADRA BPI 0.032 1.20E−28 0.417 79.2% 82.9% ITGAM 3.00E−15
    ITGAL VEGFB HLADRA 1.60E−14 1.20E−39 0.544 83.1% 86.2% ITGAL 2.00E−28
    ITGAL VEGFB BCL2 4.70E−07 2.20E−32 0.452 80.0% 82.2% ITGAL 1.20E−15
    ITGAL VEGFB CASP9 5.80E−05 2.20E−30 0.427 80.0% 80.3% ITGAL 2.00E−10
    ITGAL VEGFB NFKB1 0.0021 6.10E−29 0.41 76.9% 80.9% ITGAL 4.90E−13
    ITGAL VEGFB IFI16 0.0077 1.90E−28 0.402 76.9% 81.6% ITGAL 3.70E−21
    ITGAL VEGFB CD14 0.014 3.30E−28 0.4 76.2% 81.6% ITGAL 5.30E−27
    ITGAL VEGFB NFKBIB 0.026 5.70E−28 0.397 79.2% 80.3% ITGAL 8.80E−15
    ITGAL VEGFB CCR3 0.026 5.70E−28 0.397 77.7% 80.9% ITGAL 2.50E−29
    ITGAL VEGFB PI3 0.041 8.20E−28 0.396 76.9% 81.6% ITGAL 3.10E−21
    ITGAL VEGFB ITGAM 0.043 4.00E−28 0.409 78.5% 81.6% ITGAL 3.70E−15
    CASP9 TGFBR2 HLADRA 4.60E−17 2.60E−36 0.515 83.8% 82.2%
    CASP9 TGFBR2 CCR3 0.00031 5.40E−24 0.354 80.0% 78.9%
    CASP9 TGFBR2 IFI16 0.0014 2.10E−23 0.347 78.5% 78.9%
    CASP9 TGFBR2 ITGAL 0.002 3.00E−23 0.348 74.6% 82.9%
    CASP9 TGFBR2 JUN 0.0087 1.10E−22 0.339 76.2% 79.6%
    CASP9 TGFBR2 CD4 0.018 2.10E−22 0.334 76.2% 78.3%
    CASP9 IFI16 HLADRA 1.40E−18 1.30E−36 0.506 83.1% 84.9%
    CASP9 IFI16 CD14 0.00011 4.00E−23 0.335 75.4% 77.6%
    CASP9 IFI16 CCR3 0.0009 2.80E−22 0.323 74.6% 73.7%
    CASP9 IFI16 JUN 0.0024 1.20E−21 0.326 76.9% 77.6%
    CASP9 IFI16 ITGAL 0.0027 7.50E−22 0.319 74.6% 75.0%
    CASP9 IFI16 PI3 0.0075 1.90E−21 0.314 75.4% 72.4%
    CASP9 IFI16 CD4 0.025 5.50E−21 0.307 74.6% 73.0%
  • TABLE 8
    4 gene models capable of correctly classifying MS v. Normal Subjects
    incremental incremental incremental incremental
    p-value p-value p-value p-value
    gene
    1 gene 2 gene 3 gene 4 p-value % MS % normals gene p-value gene p-value gene p-value
    CASP9 HLADRA ITGAL CCR3 0.006 85.4% 83.6% CASP9 9.00E−06 HLADRA 9.40E−21 ITGAL 3.00E−09
  • TABLE 9
    5 gene models capable of correctly classifying MS v. Normal Subjects
    incremental incremental
    p-value p-value
    gene
    1 gene 2 gene 3 gene 4 gene 5 p-value % MS % normals gene p-value
    CASP9 HLADRA ITGAL CCR3 TGFBR2 0.0015 86.9% 84.2% CASP9 6.20E−08
    incremental incremental incremental
    p-value p-value p-value
    gene p-value gene p-value gene p-value
    HLADRA 5.90E−18 ITGAL 1.60E−07 CCR3 0.0023
  • TABLE 10
    Precision Profile ™ for Inflammatory Response
    Gene Gene Accession
    Symbol Gene Name Number
    ADAM17 a disintegrin and metalloproteinase domain 17 (tumor necrosis NM_003183
    factor, alpha, converting enzyme)
    ALOX5 arachidonate 5-lipoxygenase NM_000698
    ANXA11 annexin A11 NM_001157
    APAF1 apoptotic Protease Activating Factor 1 NM_013229
    BAX BCL2-associated X protein NM_138761
    C1QA complement component 1, q subcomponent, alpha polypeptide NM_015991
    CASP1 caspase 1, apoptosis-related cysteine peptidase (interleukin 1, NM_033292
    beta, convertase)
    CASP3 caspase 3, apoptosis-related cysteine peptidase NM_004346
    CCL2 chemokine (C—C motif) ligand 2 NM_002982
    CCL3 chemokine (C—C motif) ligand 3 NM_002983
    CCL5 chemokine (C—C motif) ligand 5 NM_002985
    CCR3 chemokine (C—C motif) receptor 3 NM_001837
    CCR5 chemokine (C—C motif) receptor 5 NM_000579
    CD14 CD14 antigen NM_000591
    CD19 CD19 Antigen NM_001770
    CD4 CD4 antigen (p55) NM_000616
    CD86 CD86 antigen (CD28 antigen ligand 2, B7-2 antigen) NM_006889
    CD8A CD8 antigen, alpha polypeptide NM_001768
    CRP C-reactive protein, pentraxin-related NM_000567
    CSF2 colony stimulating factor 2 (granulocyte-macrophage) NM_000758
    CSF3 colony stimulating factor 3 (granulocytes) NM_000759
    CTLA4 cytotoxic T-lymphocyte-associated protein 4 NM_005214
    CXCL1 chemokine (C—X—C motif) ligand 1 (melanoma growth NM_001511
    stimulating activity, alpha)
    CXCL10 chemokine (C—X—C moif) ligand 10 NM_001565
    CXCL3 chemokine (C—X—C motif) ligand 3 NM_002090
    CXCL5 chemokine (C—X—C motif) ligand 5 NM_002994
    CXCR3 chemokine (C—X—C motif) receptor 3 NM_001504
    DPP4 Dipeptidylpeptidase 4 NM_001935
    EGR1 early growth response-1 NM_001964
    ELA2 elastase 2, neutrophil NM_001972
    FAIM3 Fas apoptotic inhibitory molecule 3 NM_005449
    FASLG Fas ligand (TNF superfamily, member 6) NM_000639
    GCLC glutamate-cysteine ligase, catalytic subunit NM_001498
    GZMB granzyme B (granzyme 2, cytotoxic T-lymphocyte-associated NM_004131
    serine esterase 1)
    HLA-DRA major histocompatibility complex, class II, DR alpha NM_019111
    HMGB1 high-mobility group box 1 NM_002128
    HMOX1 heme oxygenase (decycling) 1 NM_002133
    HSPA1A heat shock protein 70 NM_005345
    ICAM1 Intercellular adhesion molecule 1 NM_000201
    ICOS inducible T-cell co-stimulator NM_012092
    IFI16 interferon inducible protein 16, gamma NM_005531
    IFNG interferon gamma NM_000619
    IL10 interleukin 10 NM_000572
    IL12B interleukin 12 p40 NM_002187
    IL13 interleukin 13 NM_002188
    IL15 Interleukin 15 NM_000585
    IRF1 interferon regulatory factor 1 NM_002198
    IL18 interleukin 18 NM_001562
    IL18BP IL-18 Binding Protein NM_005699
    IL1A interleukin 1, alpha NM_000575
    IL1B interleukin 1, beta NM_000576
    IL1R1 interleukin 1 receptor, type I NM_000877
    IL1RN interleukin 1 receptor antagonist NM_173843
    IL2 interleukin 2 NM_000586
    IL23A interleukin 23, alpha subunit p19 NM_016584
    IL32 interleukin 32 NM_001012631
    IL4 interleukin 4 NM_000589
    IL5 interleukin 5 (colony-stimulating factor, eosinophil) NM_000879
    IL6 interleukin 6 (interferon, beta 2) NM_000600
    IL8 interleukin 8 NM_000584
    LTA lymphotoxin alpha (TNF superfamily, member 1) NM_000595
    MAP3K1 mitogen-activated protein kinase kinase kinase 1 XM_042066
    MAPK14 mitogen-activated protein kinase 14 NM_001315
    MHC2TA class II, major histocompatibility complex, transactivator NM_000246
    MIF macrophage migration inhibitory factor (glycosylation-inhibiting NM_002415
    factor)
    MMP12 matrix metallopeptidase 12 (macrophage elastase) NM_002426
    MMP8 matrix metallopeptidase 8 (neutrophil collagenase) NM_002424
    MMP9 matrix metallopeptidase 9 (gelatinase B, 92 kDa gelatinase, 92 kDa NM_004994
    type IV collagenase)
    MNDA myeloid cell nuclear differentiation antigen NM_002432
    MPO myeloperoxidase NM_000250
    MYC v-myc myelocytomatosis viral oncogene homolog (avian) NM_002467
    NFKB1 nuclear factor of kappa light polypeptide gene enhancer in B-cells NM_003998
    1 (p105)
    NOS2A nitric oxide synthase 2A (inducible, hepatocytes) NM_000625
    PLA2G2A phospholipase A2, group IIA (platelets, synovial fluid) NM_000300
    PLA2G7 phospholipase A2, group VII (platelet-activating factor NM_005084
    acetylhydrolase, plasma)
    PLAU plasminogen activator, urokinase NM_002658
    PLAUR plasminogen activator, urokinase receptor NM_002659
    PRTN3 proteinase 3 (serine proteinase, neutrophil, Wegener NM_002777
    granulomatosis autoantigen)
    PTGS2 prostaglandin-endoperoxide synthase 2 (prostaglandin G/H NM_000963
    synthase and cyclooxygenase)
    PTPRC protein tyrosile phosphatase, receptor type, C NM_002838
    PTX3 pentraxin-related gene, rapidly induced by IL-1 beta NM_002852
    SERPINA1 serine (or cysteine) proteinase inhibitor, clade A (alpha-1 NM_000295
    antiproteinase, antitrypsin), member 1
    SERPINE1 serpin peptidase inhibitor, clade E (nexin, plasminogen activator NM_000602
    inhibitor type 1), member 1
    SSI-3 suppressor of cytokine signaling 3 NM_003955
    TGFB1 transforming growth factor, beta 1 (Camurati-Engelmann disease) NM_000660
    TIMP1 tissue inhibitor of metalloproteinase 1 NM_003254
    TLR2 toll-like receptor 2 NM_003264
    TLR4 toll-like receptor 4 NM_003266
    TNF tumor necrosis factor (TNF superfamily, member 2) NM_000594
    TNFRSF13B tumor necrosis factor receptor superfamily, member 13B NM_012452
    TNFRSF17 tumor necrosis factor receptor superfamily, member 17 NM_001192
    TNFRSF1A tumor necrosis factor receptor superfamily, member 1A NM_001065
    TNFSF13B Tumor necrosis factor (ligand) superfamily, member 13b NM_006573
    TNFSF5 CD40 ligand (TNF superfamily, member 5, hyper-IgM syndrome) NM_000074
    TXNRD1 thioredoxin reductase NM_003330
    VEGF vascular endothelial growth factor NM_003376

Claims (35)

1. A method for predicting an increased risk to an adverse effect from anti-TNF therapy in a subject, based on a sample from the subject, the sample providing a source of RNAs, said method comprising:
a) assessing a profile data set of a plurality of members, each member being a quantitative measure of the amount of a distinct RNA constituent in a panel of constituents selected so that measurement of the constituents enables characterization of the presumptive signs of multiple sclerosis or an inflammatory condition related to multiple sclerosis, wherein such measure for each constituent is obtained under measurement conditions that are substantially repeatable to produce a patient data set; and
b) comparing the patient data set to a baseline profile data set, wherein the baseline profile data set is related to said multiple sclerosis or inflammatory condition related to multiple sclerosis;
wherein a similarity between the patient data set and the baseline profile data set indicates a risk of an adverse effect from anti-TNF therapy in the subject.
2. The method of claim 1, wherein said subject has an inflammatory condition selected from the group consisting of rheumatoid arthritis, psoriasis, ankylosing spondylitis, psoriatic arthritis and Crohn's diseases.
3. The method of claim 2, wherein said sample is obtained prior to administering an anti-TNF therapeutic to the subject.
4. The method of claim 2, wherein said sample is obtained during the course of anti-TNF therapy.
5. The method of claim 2, wherein is obtained after administration of an anti-TNF therapeutic to the subject.
6. The method of claim 1, wherein the panel comprises 10 or fewer constituents.
7. The method of claim 1, wherein the panel comprises 5 or fewer constituents.
8. The method of claim 1, wherein the panel comprises 2 constituents,
9. The method of claim 1, wherein the panel of constituents distinguishes from a normal and a MS-diagnosed subject with at least 75% accuracy.
10. The method of claim 1, wherein the panel includes ITGAM.
11. The method of claim 10, wherein the panel further includes CD4 and MMP9.
12. The method of claim 10, wherein the panel further includes ITGA4 and MMP9.
13. A method according to claim 12, wherein the panel further includes CALCA.
14. A method according to claim 13, wherein the panel further includes CXCR3.
15. A method according to claim 12, wherein the panel further includes NFKB1B.
16. A method according to claim 15, wherein the panel further includes CXCR3.
17. The method of claim 1, wherein the panel includes HLADRA.
18. The method of claim 1, wherein the panel includes two or more constituents from Table 4 or 10.
19. A method for predicting an increased risk of an adverse effect from anti-TNF therapy in a subject, based on a sample from the subject, the sample providing a source of RNAs, said method comprising:
a) determining a quantitative measure of the amount of at least one constituent of Table 4 or 10 as a distinct RNA constituent, wherein such measure is obtained under measurement conditions that are substantially repeatable to produce a patient data set;
b) comparing the patient data set to a baseline profile data set, wherein the baseline profile data set is related to said multiple sclerosis or inflammatory condition related to multiple sclerosis;
wherein a similarity between the patient data set and the baseline profile data set indicates a risk of an adverse effect from anti-TNF therapy in the subject.
20. The method of claim 19, wherein said constituent is HLDRA.
21. The method of claim 20, further comprising determining a quantitative measure of at least one constituent selected from the group consisting of ITGAL, CASP9, NFKBIB, STAT2, NFKB1, ITGAM, ITGAL, CD4, IL1B, HSPA1A, ICAM1, IFI16, or TGFBR2.
22. The method of claim 19, wherein said constituent is CASP9.
23. The method of claim 22, further comprising determining a quantitative measure of at least one constituent selected from the group consisting of VEGFB, CD14, or JUN.
24. The method of claim 19, wherein said constituent is ITGAL
25. The method of claim 24, further comprising determining a quantitative measure of at least one constituent selected from the group consisting of P13, ITGAM, TGFBR2
26. The method of claim 19, wherein said constituent is STAT3
27. The method of claim 26, further comprising determining a qualitative measure of CD14.
28. The method of claim 19, wherein the constituents distinguish from a normal and a MS-diagnosed subject with at least 75% accuracy.
29. The method of claim 19, comprising determining a qualitative measure of three constituents in any combination shown on Table 7.
30. A method for determining a profile data set according to claim 1 or 19, wherein the measurement conditions that are substantially repeatable are within a degree of repeatability of better than five percent.
31. A method of claim 1, or 19, wherein the measurement conditions that are substantially repeatable are within a degree of repeatability of better than three percent.
32. A method of claim 1, or 19, wherein efficiencies of amplification for all constituents are substantially similar.
33. A method of claim 1 or 19, wherein the efficiency of amplification for all constituents is within two percent.
34. A method of claim 1, or 19, wherein the efficiency of amplification for all constituents is less than one percent.
35. A method of claim 1 or 19 wherein the sample is selected from the group consisting of blood, a blood fraction, body fluid, a population of cells and tissue from the subject.
US11/827,892 1999-06-28 2007-07-13 Gene expression profiling for identification, monitoring and treatment of multiple sclerosis Abandoned US20080183395A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/827,892 US20080183395A1 (en) 1999-06-28 2007-07-13 Gene expression profiling for identification, monitoring and treatment of multiple sclerosis

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US14154299P 1999-06-28 1999-06-28
US19552200P 2000-04-07 2000-04-07
US60558100A 2000-06-28 2000-06-28
US09/821,850 US6692916B2 (en) 1999-06-28 2001-03-29 Systems and methods for characterizing a biological condition or agent using precision gene expression profiles
US10/291,225 US6960439B2 (en) 1999-06-28 2002-11-08 Identification, monitoring and treatment of disease and characterization of biological condition using gene expression profiles
US43525702P 2002-12-19 2002-12-19
US10/742,458 US20050060101A1 (en) 1999-06-28 2003-12-19 Systems and methods for characterizing a biological condition or agent using precision gene expression profiles
US11/155,930 US20060115826A1 (en) 1999-06-28 2005-06-16 Gene expression profiling for identification monitoring and treatment of multiple sclerosis
US11/827,892 US20080183395A1 (en) 1999-06-28 2007-07-13 Gene expression profiling for identification, monitoring and treatment of multiple sclerosis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/454,533 Continuation-In-Part US20070014650A1 (en) 2005-07-12 2006-06-16 Fastener retention mechanism

Publications (1)

Publication Number Publication Date
US20080183395A1 true US20080183395A1 (en) 2008-07-31

Family

ID=57046856

Family Applications (1)

Application Number Title Priority Date Filing Date
US11/827,892 Abandoned US20080183395A1 (en) 1999-06-28 2007-07-13 Gene expression profiling for identification, monitoring and treatment of multiple sclerosis

Country Status (1)

Country Link
US (1) US20080183395A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100055722A1 (en) * 2007-11-30 2010-03-04 Nayak Ramesh C Methods of Detecting A Neurological Condition Via Analysis of Circulating Phagocytes
US20110129857A1 (en) * 2009-11-27 2011-06-02 Msdx, Inc. Methods Of Detecting Or Monitoring Activity Of An Inflammatory Condition Or Neurodegenerative Condition
US8673309B2 (en) 2011-02-11 2014-03-18 Msdx, Inc. Methods for measuring high molecular weight complexes of fibrinogen with fibronectin and fibulin-1
US9128101B2 (en) 2010-03-01 2015-09-08 Caris Life Sciences Switzerland Holdings Gmbh Biomarkers for theranostics
US9469876B2 (en) 2010-04-06 2016-10-18 Caris Life Sciences Switzerland Holdings Gmbh Circulating biomarkers for metastatic prostate cancer
WO2016123329A3 (en) * 2015-01-28 2016-11-03 Genentech, Inc. Gene expression markers and treatment of multiple sclerosis
US11424031B2 (en) * 2016-09-14 2022-08-23 Hoffmann-La Roche Inc. Digital biomarkers for progressing MS

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5506145A (en) * 1994-12-02 1996-04-09 Bull; Brian S. Determination of an individual's inflammation index from whole blood fibrinogen and hematocrit or hemoglobin measurements
US5643765A (en) * 1993-04-06 1997-07-01 University Of Rochester Method for quantitative measurement of gene expression using multiplex competitive reverse transcriptase-polymerase chain reaction
US5811231A (en) * 1993-01-21 1998-09-22 Pres. And Fellows Of Harvard College Methods and kits for eukaryotic gene profiling
US5866330A (en) * 1995-09-12 1999-02-02 The Johns Hopkins University School Of Medicine Method for serial analysis of gene expression
US5968784A (en) * 1997-01-15 1999-10-19 Chugai Pharmaceutical Co., Ltd. Method for analyzing quantitative expression of genes
US5994076A (en) * 1997-05-21 1999-11-30 Clontech Laboratories, Inc. Methods of assaying differential expression
US6132969A (en) * 1998-06-19 2000-10-17 Rosetta Inpharmatics, Inc. Methods for testing biological network models
US6146828A (en) * 1996-08-14 2000-11-14 Exact Laboratories, Inc. Methods for detecting differences in RNA expression levels and uses therefor
US6185561B1 (en) * 1998-09-17 2001-02-06 Affymetrix, Inc. Method and apparatus for providing and expression data mining database
US6203988B1 (en) * 1998-01-14 2001-03-20 Hitachi, Ltd. DNA fragment preparation method for gene expression profiling
US6232065B1 (en) * 1998-05-06 2001-05-15 Case Western Reserve University Analysis of gene family expression
US6607879B1 (en) * 1998-02-09 2003-08-19 Incyte Corporation Compositions for the detection of blood cell and immunological response gene expression
US6692916B2 (en) * 1999-06-28 2004-02-17 Source Precision Medicine, Inc. Systems and methods for characterizing a biological condition or agent using precision gene expression profiles
US6759197B2 (en) * 2000-03-31 2004-07-06 Sir Mortimer B. Davis -- Jewish General Hospital Microchip arrays of regulatory genes
US7130746B2 (en) * 1998-05-08 2006-10-31 Rosetta Inpharmatics Llc Computer systems and computer programs for determining protein activity levels using gene expression profiles

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5811231A (en) * 1993-01-21 1998-09-22 Pres. And Fellows Of Harvard College Methods and kits for eukaryotic gene profiling
US5643765A (en) * 1993-04-06 1997-07-01 University Of Rochester Method for quantitative measurement of gene expression using multiplex competitive reverse transcriptase-polymerase chain reaction
US5506145A (en) * 1994-12-02 1996-04-09 Bull; Brian S. Determination of an individual's inflammation index from whole blood fibrinogen and hematocrit or hemoglobin measurements
US5866330A (en) * 1995-09-12 1999-02-02 The Johns Hopkins University School Of Medicine Method for serial analysis of gene expression
US6146828A (en) * 1996-08-14 2000-11-14 Exact Laboratories, Inc. Methods for detecting differences in RNA expression levels and uses therefor
US5968784A (en) * 1997-01-15 1999-10-19 Chugai Pharmaceutical Co., Ltd. Method for analyzing quantitative expression of genes
US5994076A (en) * 1997-05-21 1999-11-30 Clontech Laboratories, Inc. Methods of assaying differential expression
US6203988B1 (en) * 1998-01-14 2001-03-20 Hitachi, Ltd. DNA fragment preparation method for gene expression profiling
US6607879B1 (en) * 1998-02-09 2003-08-19 Incyte Corporation Compositions for the detection of blood cell and immunological response gene expression
US6232065B1 (en) * 1998-05-06 2001-05-15 Case Western Reserve University Analysis of gene family expression
US7130746B2 (en) * 1998-05-08 2006-10-31 Rosetta Inpharmatics Llc Computer systems and computer programs for determining protein activity levels using gene expression profiles
US6132969A (en) * 1998-06-19 2000-10-17 Rosetta Inpharmatics, Inc. Methods for testing biological network models
US6185561B1 (en) * 1998-09-17 2001-02-06 Affymetrix, Inc. Method and apparatus for providing and expression data mining database
US6692916B2 (en) * 1999-06-28 2004-02-17 Source Precision Medicine, Inc. Systems and methods for characterizing a biological condition or agent using precision gene expression profiles
US6759197B2 (en) * 2000-03-31 2004-07-06 Sir Mortimer B. Davis -- Jewish General Hospital Microchip arrays of regulatory genes

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100055722A1 (en) * 2007-11-30 2010-03-04 Nayak Ramesh C Methods of Detecting A Neurological Condition Via Analysis of Circulating Phagocytes
US8506933B2 (en) 2007-11-30 2013-08-13 Msdx, Inc. Methods of detecting a neurological condition via analysis of circulating phagocytes
WO2010063009A1 (en) * 2008-11-28 2010-06-03 Nayak Ramesh C Methods of detecting a neurological condition via analysis of circulating phagocytes
US20110129857A1 (en) * 2009-11-27 2011-06-02 Msdx, Inc. Methods Of Detecting Or Monitoring Activity Of An Inflammatory Condition Or Neurodegenerative Condition
US9128101B2 (en) 2010-03-01 2015-09-08 Caris Life Sciences Switzerland Holdings Gmbh Biomarkers for theranostics
US9469876B2 (en) 2010-04-06 2016-10-18 Caris Life Sciences Switzerland Holdings Gmbh Circulating biomarkers for metastatic prostate cancer
US8673309B2 (en) 2011-02-11 2014-03-18 Msdx, Inc. Methods for measuring high molecular weight complexes of fibrinogen with fibronectin and fibulin-1
WO2016123329A3 (en) * 2015-01-28 2016-11-03 Genentech, Inc. Gene expression markers and treatment of multiple sclerosis
JP2018506275A (en) * 2015-01-28 2018-03-08 ジェネンテック, インコーポレイテッド Gene expression markers and treatment of multiple sclerosis
US11236391B2 (en) 2015-01-28 2022-02-01 Genentech, Inc. Gene expression markers and treatment of multiple sclerosis
US11424031B2 (en) * 2016-09-14 2022-08-23 Hoffmann-La Roche Inc. Digital biomarkers for progressing MS

Similar Documents

Publication Publication Date Title
US7935482B2 (en) Gene expression profiling for identification monitoring and treatment of rheumatoid arthritis
US20080233573A1 (en) Gene expression profiling for identification, monitoring and treatment of transplant rejection
US9809854B2 (en) Biomarkers for disease activity and clinical manifestations systemic lupus erythematosus
US20080183395A1 (en) Gene expression profiling for identification, monitoring and treatment of multiple sclerosis
WO2003040404A1 (en) Identification, monitoring and treatment of disease and characterization of biological condition using gene expression profiles
US20130095099A1 (en) Genes and genes combinations predictive of early response or non response of subjects suffering from inflammatory disease to cytokine targeting drugs (cytd)
EP2069533B1 (en) Gene expression profiling for identification, monitoring and treatment of multiple sclerosis
EP2668287B1 (en) Genes and genes combinations based on gene mknk1 predictive of early response or non response of subjects suffering from rheumatoid arthritis to tnf-alpha blocking monoclonal antibody
CA2672961A1 (en) Gene expression profiling for identification, monitoring, and treatment of ocular disease
US20110300542A1 (en) Gene Expression Profiling For Identification, Monitoring And Treatment Of Multiple Sclerosis
US20100285458A1 (en) Gene Expression Profiling for Identification, Monitoring, and Treatment of Lupus Erythematosus
CA2728537A1 (en) Gene expression profiling for identification, monitoring, and treatment of osteoarthritis
EP2062981B1 (en) Gene expression profiling for identification and monitoring of multiple sclerosis
US20060115826A1 (en) Gene expression profiling for identification monitoring and treatment of multiple sclerosis
AU2003303238B2 (en) Identification, monitoring and treatment of infectious disease and characterization of inflammatory conditions related to infectious disease using gene expression profiles
EP2024514B1 (en) Assessment of effect of an agent on a human biological condition using rodent gene expression panels
EP3758005A1 (en) Identification of the cellular function of an active nfkb pathway

Legal Events

Date Code Title Description
AS Assignment

Owner name: SOURCE PRECISION MEDICINE, INC. D/B/A SOURCE MDX,

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BEVILACQUA, MICHAEL;TRYON, VICTOR;BANKAITIS-DAVIS, DANUTE;AND OTHERS;REEL/FRAME:023614/0913;SIGNING DATES FROM 20000327 TO 20080522

AS Assignment

Owner name: LIFE TECHNOLOGIES CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SOURCE PRECISION MEDICINE, INC. D/B/A SOURCE MDX;REEL/FRAME:026238/0857

Effective date: 20110505

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION